input
stringlengths 28k
164k
| pubmedid
stringlengths 8
8
| output
stringlengths 321
41.9k
|
|---|---|---|
Predicting Essential Components of Signal
Transduction Networks: A Dynamic Model
of Guard Cell Abscisic Acid Signaling
Song Li1, Sarah M. Assmann1, Re´ka Albert2*
1 Biology Department, Pennsylvania State University, University Park, Pennsylvania, United States of America, 2 Physics Department, Pennsylvania State University, University
Park, Pennsylvania, United States of America
Plants both lose water and take in carbon dioxide through microscopic stomatal pores, each of which is regulated by a
surrounding pair of guard cells. During drought, the plant hormone abscisic acid (ABA) inhibits stomatal opening and
promotes stomatal closure, thereby promoting water conservation. Dozens of cellular components have been
identified to function in ABA regulation of guard cell volume and thus of stomatal aperture, but a dynamic description
is still not available for this complex process. Here we synthesize experimental results into a consistent guard cell
signal transduction network for ABA-induced stomatal closure, and develop a dynamic model of this process. Our
model captures the regulation of more than 40 identified network components, and accords well with previous
experimental results at both the pathway and whole-cell physiological level. By simulating gene disruptions and
pharmacological interventions we find that the network is robust against a significant fraction of possible
perturbations. Our analysis reveals the novel predictions that the disruption of membrane depolarizability, anion
efflux, actin cytoskeleton reorganization, cytosolic pH increase, the phosphatidic acid pathway, or Kþ efflux through
slowly activating Kþ channels at the plasma membrane lead to the strongest reduction in ABA responsiveness. Initial
experimental analysis assessing ABA-induced stomatal closure in the presence of cytosolic pH clamp imposed by the
weak acid butyrate is consistent with model prediction. Simulations of stomatal response as derived from our model
provide an efficient tool for the identification of candidate manipulations that have the best chance of conferring
increased drought stress tolerance and for the prioritization of future wet bench analyses. Our method can be readily
applied to other biological signaling networks to identify key regulatory components in systems where quantitative
information is limited.
Citation: Li S, Assmann SM, Albert R (2006) Predicting essential components of signal transduction networks: A dynamic model of guard cell abscisic acid signaling. PLoS Biol
4(10): e312. DOI: 10.1371/journal.pbio.0040312
Introduction
One central challenge of systems biology is the distillation
of systems level information into applications such as drug
discovery in biomedicine or genetic modification of crops. In
terms of applications it is important and practical that we
identify the subset of key components and regulatory
interactions whose perturbation or tuning leads to significant
functional changes (e.g., changes in a crop’s fitness under
environmental stress or changes in the state of malfunction-
ing cells, thereby combating disease). Mathematical modeling
can assist in this process by integrating the behavior of
multiple components into a comprehensive model that goes
beyond human intuition, and also by addressing questions
that are not yet accessible to experimental analysis.
In recent years, theoretical and computational analysis of
biochemical networks has been successfully applied to well-
defined metabolic pathways, signal transduction, and gene
regulatory networks [1–3]. In parallel, high-throughput
experimental methods have enabled the construction of
genome-scale maps of transcription factor–DNA and pro-
tein–protein interactions [4,5]. The former are quantitative,
dynamic descriptions of experimentally well-studied cellular
pathways with relatively few components, while the latter are
static maps of potential interactions with no information
about their timing or kinetics. Here we introduce a novel
approach that stands in the middle ground of the above-
mentioned methods by incorporating the synthesis and
dynamic modeling of complex cellular networks that contain
diverse, yet only qualitatively known regulatory interactions.
We develop a mathematical model of a highly complex
cellular signaling network and explore the extent to which
the network topology determines the dynamic behavior of
the system. We choose to examine signal transduction in
plant guard cells for two reasons. First, guard cells are central
components in control of plant water balance, and better
Academic Editor: Joanne Chory, The Salk Institute for Biological Studies, United
States of America
Received April 3, 2006; Accepted July 21, 2006; Published September 12, 2006
DOI: 10.1371/journal.pbio.0040312
Copyright: 2006 Li et al. This is an open-access article distributed under the
terms of the Creative Commons Attribution License, which permits unrestricted
use, distribution, and reproduction in any medium, provided the original author
and source are credited.
Abbreviations: ABA, abscisic acid; PP2C, protein phosphatase 2C; Atrboh, NADPH
oxidase; Ca2þ
c , cytosolic Ca2þ increase; CaIM, Ca2þ influx across the plasma
membrane; CIS, Ca2þ influx to the cytosol from intracellular stores; CPC, cumulative
percentage of closure; GCR1, G protein–coupled receptor 1; GPA1, heterotrimeric G
protein a subunit 1; KAP, Kþ efflux through rapidly activating Kþ channels (AP
channels) at the plasma membrane; KOUT, Kþ efflux through slowly activating
outwardly-rectifying Kþ channels at the plasma membrane; NO, nitric oxide; NOS,
nitric oxide synthase; PA, phosphatidic acid; ROS, reactive oxygen species
* To whom correspondence should be addressed. E-mail: ralbert@phys.psu.edu
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1732
PLoS BIOLOGY
understanding of their regulation is important for the goal of
engineering crops with improved drought tolerance. Second,
abscisic acid (ABA) signal transduction in guard cells is one of
the best characterized signaling systems in plants: more than
20 components, including signal transduction proteins,
secondary metabolites, and ion channels, have been shown
to participate in ABA-induced stomatal closure. ABA induces
guard cell shrinkage and stomatal closure via two major
secondary messengers, cytosolic Ca2þ (Ca2þ
c) and cytosolic pH
(pHc). A number of signaling proteins and secondary
messengers have been identified as regulators of Ca2þ influx
from outside the cell or Ca2þ release from internal stores; the
downstream components responding to Ca2þ are certain
vacuolar and plasma membrane Kþ permeable channels, and
anion channels in the plasma membrane [6,7]. Increases in
cytosolic pH promote the opening of anion efflux channels
and enhance the opening of voltage-activated outward Kþ
channels in the plasma membrane [8–10]. Stomatal closure is
caused by osmotically driven cell volume changes induced by
both Kþ and anion efflux through plasma membrane–
localized channels. Despite the wealth of information that
has been collected regarding ABA signal transduction, the
majority of the regulatory relationships are known only
qualitatively and are studied in relative isolation, without
considering their possible feedback or crosstalk with other
pathways. Therefore, in order to synthesize this rich knowl-
edge, one needs to assemble the information on regulatory
mechanisms involved in ABA-induced stomatal closure into a
system-level regulatory network that is consistent with
experimental observations. Clearly, it is difficult to assemble
the network and predict the dynamics of this system from
human intuition alone, and thus theoretical tools are needed.
We synthesize the experimental information available
about the components and processes involved in ABA-
induced stomatal closure into a comprehensive network,
and study the topology of paths between signal and response.
To capture the dynamics of information flow in this network
we express synergy between pathways as combinatorial rules
for the regulation of each node, and formulate a dynamic
model of ABA-induced closure. Both in silico and in initial
experimental analysis, we study the resilience of the signaling
network to disruptions. We systematically sample functional
and dynamic perturbations in network components and
uncover a rich dynamic repertoire ranging from ABA
hypersensitivity to complete insensitivity. Our model is
validated by its agreement with prior experimental results,
and yields a variety of novel predictions that provide targets
on which further experimental analysis should focus. To our
knowledge, this is one of the most complex biological
networks ever modeled in a dynamical fashion.
Results
Extraction and Organization of Data from the Literature
We focus on ABA induction of stomatal closure, rather
than ABA inhibition of stomatal opening, because these two
processes, although related, exhibit distinct mechanisms, and
there is substantially more information on the former process
than on the latter in the literature. Experimental information
about the involvement of a specific component in ABA-
induced stomatal closure can be partitioned into three
categories. First, biochemical evidence provides information
on enzymatic activity or protein–protein interactions. For
example, the putative G protein–coupled receptor 1 (GCR1)
can physically interact with the heterotrimeric G protein a
component 1 (GPA1) as supported by split-ubiquitin and
coimmunoprecipitation experiments [11]. Second, genetic
evidence of differential responses to a stimulus in wild-type
plants versus mutant plants implicates the product of the
mutated gene in the signal transduction process. For
example, the ethyl methanesulfonate–generated ost1 mutant
is less sensitive to ABA; thus, one can infer that the OST1
protein is a part of the ABA signaling cascade [12]. Third,
pharmacological experiments, in which a chemical is used
either to mimic the elimination of a particular component, or
to exogenously provide a certain component, can lead to
similar inferences. For example, a nitric oxide (NO) scavenger
inhibits ABA-induced closure, while a NO donor promotes
stomatal closure; thus, NO is a part of the ABA network [13].
The last two types of inference do not give direct interactions
but correspond to pathways and pathway regulation. The
existing theoretical literature on signaling is focused on
networks where the first category of information is known,
along with the kinetics of each interaction. However, the
availability of such detailed knowledge is very much the
exception rather than the norm in the experimental
literature. Here we propose a novel method of representing
qualitative and incomplete experimental information and
integrating it into a consistent signal transduction network.
First, we distill experimental conclusions into qualitative
regulatory relationships between cellular components (signal-
ing proteins, metabolites, ion channels) and processes. For
example, the evidence regarding OST1 and NO is summar-
ized as both OST1 and NO promoting ABA-induced stomatal
closure. We distinguish between positive and negative
regulation by using the verbs ‘‘promote’’ and ‘‘inhibit,’’
represented graphically as ‘‘!’’ and ‘‘—j,’’ respectively, and
quantify the severity of the effect by the qualifier ‘‘partial.’’ A
partial promoter’s (inhibitor’s) loss has less severe effects than
the loss of a promoter (inhibitor), most probably due to other
regulatory effects on the target node. Using these relations,
we construct a database that contains more than 140 entries
and is derived from more than 50 literature citations on ABA
regulation of stomatal closure (Table S1). A number of entries
in the database correspond to a component-to-component
relationship, such as ‘‘A promotes B,’’ which is mostly
obtained by pharmacological experiments (e.g., applying A
causes B response). However, the majority of the entries
belong to the two categories of indirect inference described
above, and are of the type ‘‘C promotes the process (A
promotes B).’’ This kind of information can be obtained from
both genetic and pharmacological experiments (e.g., disrupt-
ing C causes less A-induced B response, or applying C and A
simultaneously causes a stronger B response than applying A
only). There are a few instances of documented independence
of two cellular components, which we identify with the
qualifier ‘‘no relationship.’’ Most of the information is
derived from the model species Arabidopsis thaliana, but data
from other species, mostly Vicia faba, are also included where
comparable information from Arabidopsis thaliana is lacking.
Assembly of the ABA Signal Transduction Network
To synthesize all this information into a consistent
network, we need to determine how the different pathways
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1733
Model of Guard Cell ABA Signaling
suggested by experiments fit together (i.e., we need to find the
pathways’ branching and crossing points). We develop a set of
rules compatible with intuitive inference, aiming to deter-
mine the sparsest graph consistent with all experimental
observations. We summarize the most important rules in
Figure 1; in the following we give examples for their
application.
If A ! B and C ! process (A ! B), where A ! B is not a
biochemical reaction such as an enzyme catalyzed reaction or
protein–protein/small molecule interaction, we assume that C
is acting on an intermediary node (IN) of the A–B pathway.
This IN could be an intermediate protein complex, protein–
small molecule complex, or multiple complexes (see Figure 1,
panel 1). For example, ABA ! closure, and NO synthase
(NOS) ! process (ABA ! closure); therefore, ABA ! IN !
closure, NOS ! IN. If A ! B is a direct process such as a
biochemical reaction or a protein–protein interaction, we
assume that C ! process (A ! B) corresponds to C ! A ! B.
A ! B and C ! process (A ! B) can be transformed to A
! C ! B if A ! C is also documented. This means that the
simplest explanation is to identify the putative intermediary
node with C. For example, ABA ! NOS, and NOS ! process
(ABA ! NO) are experimentally verified and NOS is an
enzyme producing NO, therefore, we infer ABA ! NOS !
NO (see Figure 1, panel 2).
A rule similar to rule 1 applies to inhibitory interactions
(denoted by —j); however, in the case of A —j B, and C —j
process (A —j B), the logically correct representation is: A !
IN —j B, C —j IN (see Figure 1, panel 3).
The above rules constitute a heuristic algorithm for first
expanding the network wherever the experimental relation-
ships are known to be indirect, and second, minimizing the
uncertainty of the network by filtering synonymous relation-
ships. Mathematically, this algorithm is related to the
problem of finding the minimum transitive reduction of a
graph (i.e., for finding the sparsest subgraph with the same
reachability relationships as the original) [14]; however, it
differs from previously used algorithms by the fact that the
edges can have one of two signs (activating and inhibitory),
and edges corresponding to direct interactions are main-
tained.
In the reconstructed network, given in Figure 2, the
network input is ABA and the output is the node ‘‘Closure.’’
The small black filled circles represent putative intermediary
nodes mediating indirect regulatory interactions. The edges
(lines) of the network represent interactions and processes
between two components (nodes); an arrowhead at the end of
an edge represents activation, and a short segment at the end
of an edge signifies inhibition. Edges that signify interactions
derived from species other than Arabidopsis are colored light
blue. We indicate two inferred negative feedback loops on
S1P and pHc (see below) by dashed light blue lines. Nodes
involved in the same metabolic reaction or protein complex
are bordered by a gray box; only those arrows that point into
or out of the box signify information flow (signal trans-
duction). Some of the edges on Figure 2 are not explicitly
incorporated in Table S1 because they represent general
biochemical or physical knowledge (e.g., reactions inside gray
boxes or depolarization caused by anion efflux).
A brief biological description of this reconstructed net-
work (Figure 2) is as follows. ABA induces guard cell
shrinkage and stomatal closure via two major secondary
messengers, Ca2þ
c and pHc. Two mechanisms of Ca2þ
c
increase have been identified: Ca2þ influx from outside the
cell and Ca2þ release from internal stores. Ca2þ can be
released from stores by InsP3 [15] and InsP6 [16], both of
which are synthesized in response to ABA, or by cADPR and
cGMP [17], whose upstream signaling molecule, NO [13,18], is
indirectly activated by ABA. Opening of channels mediating
Ca2þ influx is mainly stimulated by reactive oxygen species
(ROS) [19], and we reconstruct two ABA-ROS pathways
involving OST1 [12] and GPA1 (L. Perfus-Barbeoch and S. M.
Assmann, unpublished data), respectively. Based on current
experimental evidence these two pathways are distinct, but
not independent. The downstream components responding
to Ca2þ are certain vacuolar and plasma membrane Kþ
permeable channels, and anion channels in the plasma
membrane [6,7]. The mechanism of pH control by ABA is
less clear, but it is known that pHc increases shortly after ABA
treatment [20,21]. Increases in pHc levels promote the
opening of anion efflux channels and enhance the opening
of voltage-activated outward Kþ channels in the plasma
membrane [8–10]. Stomatal closure is caused by osmotically
driven cell volume changes induced by Kþ and anion efflux
through plasma membrane-localized channels, and there is a
complex interregulation between ion flux and membrane
depolarization.
In addition to the secondary-messenger–induced pathways,
there are two less-well-studied ABA signaling pathways
involving the reorganization of the actin cytoskeleton, and
the organic anion malate. ABA inactivates the small GTPase
protein RAC1, which in turn blocks actin cytoskeleton
disruption [22], contributing to an ABA-induced actin
cytoskeleton reorganization process that is potentially Ca2þ
c
dependent [23]. In our model system, Arabidopsis, ABA
regulation of malate levels has not been described. However,
in V. faba it has been shown that ABA inhibits PEP carboxylase
and malate synthesis [24], and that ABA induces malate
breakdown [25]. In some conditions sucrose is an osmoticum
that contributes to guard cell turgor [26,27] but no
mechanisms of ABA regulation of sucrose levels have been
described.
The recessive mutant of the protein phosphatase 2C (PP2C)
Figure 1. Illustration of the Inference Rules Used in Network
Reconstruction
(1) If A ! B and C ! process (A ! B), where A ! B is not a biochemical
reaction such as an enzyme catalyzed reaction or protein-protein/small
molecule interaction, we assume that C is acting on an intermediary
node (IN) of the A–B pathway.
(2) If A ! B, A ! C, and C ! process (A ! B), where A ! B is not a direct
interaction, the most parsimonious explanation is that C is a member of
the A–B pathway, i.e. A ! C ! B.
(3) If A —j B and C —j process (A —j B), where A —j B is not a direct
interaction, we assume that C is inhibiting an intermediary node (IN) of
the A–B pathway. Note that A! IN —j B is the only logically consistent
representation of the A–B pathway.
DOI: 10.1371/journal.pbio.0040312.g001
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1734
Model of Guard Cell ABA Signaling
ABI1, abi1-1R, is hypersensitive to ABA [28,29]. ABI1 is
negatively regulated by phosphatidic acid (PA) and ROS, and
pHc can activate ABI1 [30–32]. ABI1 negatively regulates
RAC1 [22]. We hypothesize that ABI1 negatively regulates the
NADPH oxidase (Atrboh) because ABI1 negatively regulates
ROS production and Atrboh has been shown to be the
dominant producer of ROS in guard cells [33]. We also
assume that ABI1 inhibits anion efflux at the plasma
membrane, because the dominant abi1–1 mutant is known
to affect the ABA response of anion channels [34] and
because anion channels are documented key regulators of
ABA-induced stomatal closure [35]. Components functioning
downstream from ABI2 and its role in guard cell signaling are
not well established, so ABI2 is not included. The newly
isolated PP2C recessive mutants, AtP2C-HA [36] and AtPP2CA
[37], exhibit minor ABA hypersensitivity. However, their
Figure 2. Current Knowledge of Guard Cell ABA Signaling
The color of the nodes represents their function: enzymes are shown in red, signal transduction proteins are green, membrane transport–related nodes
are blue, and secondary messengers and small molecules are orange. Small black filled circles represent putative intermediary nodes mediating indirect
regulatory interactions. Arrowheads represent activation, and short perpendicular bars indicate inhibition. Light blue lines denote interactions derived
from species other than Arabidopsis; dashed light-blue lines denote inferred negative feedback loops on pHc and S1P. Nodes involved in the same
metabolic pathway or protein complex are bordered by a gray box; only those arrows that point into or out of the box signify information flow (signal
transduction).
The full names of network components corresponding to each abbreviated node label are: ABA, abscisic acid; ABI1/2, protein phosphatase 2C ABI1/2;
ABH1, mRNA cap binding protein; Actin, actin cytoskeleton reorganization; ADPRc, ADP ribose cyclase; AGB1, heterotrimeric G protein b component;
AnionEM, anion efflux at the plasma membrane; Arg, arginine; AtPP2C, protein phosphatase 2C; Atrboh, NADPH oxidase; CaIM, Ca2þ influx across the
plasma membrane; Ca2þ ATPase, Ca2þ ATPases and Ca2þ/Hþ antiporters responsible for Ca2þ efflux from the cytosol; Ca2þ
c , cytosolic Ca2þ increase;
cADPR, cyclic ADP-ribose; cGMP, cyclic GMP; CIS, Ca2þ influx to the cytosol from intracellular stores; DAG, diacylglycerol; Depolar, plasma membrane
depolarization; ERA1, farnesyl transferase ERA1; GC, guanyl cyclase; GCR1, putative G protein–coupled receptor; GPA1, heterotrimeric G protein a
subunit; GTP, guanosine 59-triphosphate; Hþ ATPase, Hþ ATPase at the plasma membrane; InsPK, inositol polyphosphate kinase; InsP3, inositol-1,4,5-
trisphosphate; InsP6, inositol hexakisphosphate; KAP, Kþ efflux through rapidly activating Kþ channels (AP channels) at the plasma membrane; KEV, Kþ
efflux from the vacuole to the cytosol; KOUT, Kþ efflux through slowly activating outwardly-rectifying Kþ channels at the plasma membrane; NADþ,
nicotinamide adenine dinucleotide; NADPH, nicotinamide adenine dinucleotide phosphate; NOS, Nitric oxide synthase; NIA12, Nitrate reductase; NO,
Nitric oxide; OST1, protein kinase open stomata 1; PA, phosphatidic acid; PC, phosphatidyl choline; PEPC, phosphoenolpyruvate carboxylase; PIP2,
phosphatidylinositol 4,5-bisphosphate; PLC, phospholipase C; PLD, phospholipase D; RAC1, small GTPase RAC1; RCN1, protein phosphatase 2A; ROP2,
small GTPase ROP2; ROP10, small GTPase ROP10; ROS, reactive oxygen species; SphK, sphingosine kinase; S1P, sphingosine-1-phosphate.
DOI: 10.1371/journal.pbio.0040312.g002
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1735
Model of Guard Cell ABA Signaling
downstream targets remain elusive; thus, we incorporate
them as a general inhibitor of closure denoted AtPP2C.
Mutation of the gene encoding the mRNA cap-binding
protein, ABH1, results in hypersensitivity of ABA-induced
Ca2þ
c elevation/oscillation and of anion efflux in plants grown
under some environmental conditions [38,39]. We assume an
inhibitory effect of ABH1 on Ca2þ influx across the plasma
membrane (CaIM), which can explain both of these effects
due to the Ca2þ regulation of anion efflux. Since the abh1
mutation affects transcript levels of some genes involved in
ABA response, this mutation may also affect ABA sensitivity
by altering gene expression rather than by regulation of the
rapid signaling events on which our network focuses.
Mutations in the gene encoding the farnesyl transferase
ERA1 or the gene encoding GCR1 also lead to hypersensitive
ABA-induced closure; ERA1 has been shown to negatively
regulate CaIM and anion efflux [40,41], whereas GCR1 has
been shown to be interact with GPA1 [11]. We assume that
ERA1 negatively regulates CaIM and GCR1 negatively
regulates GPA1.
Another assumption in the network is that the protein
phosphatase RCN1/PP2A regulates nitrate reductase (NIA12)
activity as observed in spinach leaf tissue; this is expected to
be a well-conserved mechanism due to the high sequence
conservation of NIA-PP2A regulatory domains [42]. Figure 2
contains two putative autoregulatory negative feedback loops
acting on S1P and pHc, respectively. The existence of
feedback regulation can be inferred from the published
timecourse measurements of S1P [43] and pHc [21]—both
indicating a fast increase in response to ABA, then a
decrease—but the mediators are currently unknown. The
assembled network is consistent with our biological knowl-
edge with minimal additional assumptions, and it will serve as
the starting point for the graph analysis and dynamic
modeling described in the following sections.
Modeling ABA Signal Transduction
Signaling networks can be represented as directed graphs
where the orientation of the edges reflects the direction of
information propagation (signal transduction). In a signal
transduction network there exists a clear starting point, the
node representing the signal (here, ABA), and one can follow
the paths (successions of edges) from that starting point to
the node(s) representing the output(s) of the network (here,
stomatal closure). The signal–output paths correspond to the
propagation of reactions in chemical space, and can be
thought of as pseudodynamics [44]. When only static
information is available, pseudodynamics takes into account
the graph theoretical properties of the signal transduction
network. For example, one can measure the number of nodes
or distinct network motifs that appear one, two,. . .n edges
away from the signal node. Such motifs reflect different
cellular signaling processing capabilities and provide impor-
tant insights into the biological processes under investigation.
Graph theoretical measures can also provide information
about the importance (centrality) of signal mediators [45] and
can predict the changes in path structure when nodes or
edges in the network are disrupted. These disruptions,
explored experimentally by genetic mutations, voltage-
clamping, or pharmacological interventions, can be modeled
in silico by removing the perturbed node and all its edges
from the graph [46]. The absence of nodes and edges will
disrupt the paths in the network, causing a possible increase
in the length of the shortest path between signal (ABA) and
output (closure), suggesting decreased ABA sensitivity, or in
severe cases the loss of all paths connecting input and output
(i.e., ABA insensitivity).
We find that there are several partially or completely
independent (nonoverlapping) paths between ABA and
closure. The path of pH-induced anion efflux is independent
of the paths involving changes in Ca2þ
c. Based on the current
knowledge incorporated in Figure 2, the path mediated by
malate breakdown is independent of both Ca2þ and pH
signaling. This result could change if evidence of a suggested
link between pH and malate regulation [47] is found; note
that regulation of malate synthesis in guard cells appears to
have cell-specific aspects [48]. Increase in Ca2þ
c can be
induced by several independent paths involving ROS, NO,
or InsP6. Thanks to the existence of numerous redundant
signal (ABA)–output (closure) paths, a complete disconnec-
tion of signal from output (loss of all the paths) is possible
only if four nodes, corresponding to actin reorganization,
pHc increase, malate breakdown, and membrane depolariza-
tion, are simultaneously disrupted. This indicates a remark-
able topological resilience, and suggests that functionally
redundant mechanisms can compensate for single gene
disruptions and can maintain at least partial ABA sensitivity.
However, path analysis alone cannot capture bidirectional
signal propagation and synergy (cooperativity) in living
biological systems. For example, two nonoverlapping paths
that reach the node closure could be functionally synergistic.
Using only path analysis, disruption of either path would not
be predicted to lead to a disconnection of the signal (ABA)
from the output (closure), but due to the synergy between the
two paths, the closure response may be strongly impaired if
either of the two paths is disrupted experimentally. Because
of such limitations of path analysis, we turn from path
analysis to a dynamic description.
Dynamic models have as input information (1) the
interactions and regulatory relationships between compo-
nents (i.e., the interaction network); (2) how the strength of
the interactions depends on the state of the interacting
components (i.e., the transfer functions); and (3) the initial
state of each component in the system. Given these, the
model will output the time evolution of the state of the
system (e.g., the system’s response to the presence or absence
of a given signal). Given the incomplete characterization of
the processes involved in ABA-induced stomatal closure (as is
typical of the current state of knowledge of cell signaling
cascades), we employ a qualitative modeling approach. We
assume that the state of the network nodes can have two
qualitative values: 0 (inactive/off) and 1 (active/on) [49]. These
values can also describe two conformational states of a
protein, such as closed and open states of an ion channel, or
basal and high activity for enzymes. This assumption is
necessary due to the absence of quantitative concentration or
activity information for the vast majority of the network
components. It is additionally justified by the fact that in the
case of combinatorial regulation or cooperative binding, the
input–output relationships are sigmoidal and thus can be
distilled into two discrete output states [50].
Since ‘‘stomatal closure’’ does not usually entail the
complete closure of the stomatal pore but rather a clear
decrease in the stomatal aperture, and since there is a
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1736
Model of Guard Cell ABA Signaling
significant variability in the response of individual stomata,
the threshold separating the off (0) and on (1) state of the
node ‘‘Closure’’ needs to invoke a population level descrip-
tion. We measured the stomatal aperture size distribution in
the absence of ABA or after treatment with 50 lM ABA (see
Materials and Methods). Our first observation was the
population-level heterogeneity of stomatal apertures even
in their resting condition (Figure 3A), a fact that may not be
widely appreciated when more standard presentations, such
as mean 6 standard error, are used (see Figure 3B). The
stomatal aperture distribution shifts towards smaller aper-
tures after ABA treatment, and also broadens considerably.
The latter result is inconsistent with the assumption of each
stomate changing its aperture according to a common
function that decreases with increasing ABA concentration,
and suggests considerable cell-to-cell variation in the degree
of response to ABA. Moreover, although there is a clear
difference between the most probable ‘‘open’’ (0 ABA) and
‘‘closed’’ (þ ABA) aperture sizes, there also exists an overlap
between the aperture size distribution of ‘‘open’’ and
‘‘closed’’ stomata. This result indicates the possibility of
differential and cell-autonomous stomatal responses to ABA.
In the absence of 6 ABA measurements on the same stomate,
we define the threshold of closure as a statistically significant
shift of the stomatal aperture distribution towards smaller
apertures in response to ABA signal transduction.
In our model the dynamics of state changes are governed
by logical (Boolean) rules giving the state transition of each
node given the state of its regulators (upstream nodes). We
determine the Boolean transfer function for each node based
on experimental evidence. The state of a node regulated by a
single upstream component will follow the state of its
regulator with a delay. If two or more pathways can
independently lead to a node’s activation, we combine them
with a logical ‘‘or’’ function. If two pathways cannot work
independently, we model their synergy as a logical ‘‘and’’
function. For nodes regulated by inhibitors we assume that
the necessary condition of their activation (state 1) is that the
inhibitor is inactive (state 0). As all putative intermediary
nodes of Figure 2 are regulated by a single activator, and
regulate a single downstream component, they only affect the
time delays between known nodes; for this reason we do not
explicitly incorporate intermediary nodes as components of
the dynamic model. Table 1 lists the regulatory rules of
known nodes of Figure 2; we give a detailed justification of
each rule in Text S1.
Frequently in Boolean models time is quantized into
regular intervals (timesteps), assuming that the duration of
all activation and decay processes is comparable [51]. For
generality we do not make this assumption, and in the
absence of timing or duration information we follow an
asynchronous method that allows for significant stochasticity
in process durations [52,53]. Choosing as a timestep the
longest duration required for a node to respond to a change
in the state of its regulator(s) (also called a round of update, as
each component’s state will be updated during this time
interval), the Boolean updating rules of an asynchronous
algorithm can be written as:
Sn
i ¼ BiðSmj
j ; Smk
k ; Sml
l ; ::Þ;
ð1Þ
where Si
n is the state of component i at timestep n, Bi is the
Boolean function associated with the node i and its regulators
j,k,l,.. and mj; mk; ml; :: 2 fn 1; ng, signifying that the time-
points corresponding to the last change in a input node’s
state can be in either the previous or current round of
updates.
Figure 3. Stomatal Aperture Distributions without ABA Treatment (gray
bars) and with 50 lM ABA (white bars)
(A) The x axis gives the stomatal aperture size and the y axis indicates the
fraction of stomata for which that aperture size was observed. The black
columns indicate the overlap between the 0 lM ABA and the 50 lM ABA
distributions.
(B) Classical bar plot representation of stomatal aperture for treatment
with 50 lM ABA (white bar, labeled 1) and without ABA treatment (gray
bar, labeled 2) using mean 6 standard error. This representation
provides minimal information on population structure.
DOI: 10.1371/journal.pbio.0040312.g003
Table 1. Boolean Rules Governing the States of the Known
(Named) Nodes in the Signal Transduction Network
Node
Boolean Regulatory Rule
NO
NO* ¼ NIA12 and NOS
PLC
PLC* ¼ ABA and Ca2þ
c
CaIM
CaIM* ¼ (ROS or not ERA1 or not ABH1) and not Depolar
GPA1
GPA1* ¼ (S1P or not GCR1) and AGB1
Atrboh
Atrboh* ¼ pHc and OST1 and ROP2 and not ABI1
Hþ ATPase Hþ ATPase* ¼ not ROS and not pHc and not Ca2þ
c
Malate
Malate* ¼ PEPC and not ABA and not AnionEM
RAC1
RAC1* ¼ not ABA and not ABI1
Actin
Actin* ¼ Ca2þ
c or not RAC1
ROS
ROS* ¼ ABA and PA and pHc
ABI1
ABI1* ¼ pHc and not PA and not ROS
KAP
KAP*¼ (not pHc or not Ca2þ
c) and Depolar
Ca2þ
c
Ca2þ
c*¼ (CaIM or CIS) and not Ca2þ ATPase
CIS
CIS* ¼ (cGMP and cADPR) or (InsP3 and InsP6)
AnionEM
AnionEM* ¼ ((Ca2þ
c or pHc) and not ABI1 ) or (Ca2þ
c and pHc)
KOUT
KOUT* ¼ (pHc or not ROS or not NO) and Depolar
Depolar
Depolar* ¼ KEV or AnionEM or not Hþ ATPase or not KOUT or Ca2þ
c
Closure
Closure* ¼ (KOUT or KAP ) and AnionEM and Actin and not Malate
The nomenclature of the nodes is given in the caption of Figure 2. The nodes that have
only one input are not listed to save space; a full description and justification can be
found in Text S1. The next state of the node on the left-hand side of the equation (marked
by *) is determined by the states of its effector nodes according to the function on the
right-hand side of the equation.
DOI: 10.1371/journal.pbio.0040312.t001
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1737
Model of Guard Cell ABA Signaling
The relative timing of each process is chosen randomly and
is changed after each update round such that we are sampling
equally among all possibilities (see Materials and Methods).
This approach reflects the lack of experimental data on
relative reaction speeds. The internal states of signaling
proteins and the concentrations of small molecules are not
explicitly known for each stomate, and components such as
Ca2þ
c and cell membrane potential show various states even
in a homogenous experimental setup [54,55]. Accordingly, we
sample a large number (10,000) of randomly selected initial
states for the nodes other than ABA and closure (closure is
initially set to 0), and let the system evolve either with ABA
always on (1) or ABA always off (0). We quantify the
probability of closure (equivalent to the percentage of closed
stomata in the population) by the formula
PðclosureÞt ¼
X
N
j¼l
St
closureðjÞ=N
ð2Þ
where St
closure(j) is the state of the node ‘‘Closure’’ at time t in
the jth simulation and N is the total number of simulations, in
our case 10,000. We illustrate the main steps of our
simulation method in Figure 4.
As shown in Figure 5, in eight steps, the system shows
complete closure in response to ABA. In contrast, without
ABA, although some initial states lead to closure at the
beginning, within six steps the probability of closure
approaches 0. Initial theoretical analysis of the attractors
(stable behaviors) of this nonlinear dynamic system confirms
that when given a constant ABA ¼ 1 input, the majority of
nodes will approach a steady state value within three to eight
steps. This steady-state value does not depend on the initial
conditions. For example, OST1, PLC, and InsPK stabilize in
the on state, and PEPC settles into the off state within the first
timestep when ABA is consistently on. The exception is a set
of 12 nodes, including Ca2þ
c, Ca2þ ATPase, NO, Kþ efflux
from the vacuole to the cytosol, and Kþ efflux through rapidly
Figure 4. Schematic Illustration of Our Modeling Methodology and of the Probability of Closure
In this four-node network example, node A is the input (as ABA is the input of the ABA signal transduction network), and node D is the output
(corresponding to the node ‘‘Closure’’ in the ABA signal transduction network). The nodes’ states are indicated by the shading of their symbols: open
symbols represent the off (0) state and filled symbols signify the on (1) state. To indicate the connection between this example and ABA-induced
closure, we associate D ¼ off (0) with a picture of an open stomate, and D ¼ on (1) with a picture of a closed stomate. The Boolean transfer functions of
this network are A* ¼ 1, B* ¼ A, C* ¼ A, D* ¼ B and C (i.e., node A is on commencing immediately after the initial condition, the next states of nodes B
and C are determined by A, and D is on only when both B and C are on).
(A) The first column represents the networks’ initial states; the input and output are not on, but some of the components in the network are randomly
activated (e.g., middle row, node B). The input node A turns on right after initialization, signifying the initiation of the ABA signal. The next three
columns in (A) represent the network’s intermediary states during a sequential update of the nodes B, C, and D, where the updated node is given as a
gray label above the gray arrow corresponding to the state transition. This sequence of three transitions represents a round of updates from timestep 1
(second column) to timestep 2 (last column). Out of a total of 22 3 3! ¼ 24 possible different normal responses, two sketches of normal responses are
shown in the top two rows. The bottom row illustrates a case in which one node (shown as a square) is disrupted (knocked out) and cannot be
regulated or regulate downstream nodes (indicated as dashed edges).
(B) The probability of closure indicates the fraction of simulations where the output D ¼ 1 is reached in each timestep; thus, in this illustration the
probability of closure for the normal response (circles) increases from 0% at time step 1 to 100% at timestep 2. The knockout mutant’s probability of
closure (squares) is 0% at both time steps.
DOI: 10.1371/journal.pbio.0040312.g004
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1738
Model of Guard Cell ABA Signaling
activating Kþ channels (AP channels) at the plasma membrane
(KAP), whose attractors are limit cycles (oscillations) accord-
ing to the model. Ca2þ
c oscillations have indeed been
observed experimentally [56,57]; no time course measure-
ments have been reported in the literature for the other
components, so it is unknown whether they oscillate or not.
We identified four subsets of behaviors for these nodes—
distinguished by different positions on the limit cycle—
depending on the initial conditions and relative process
durations. Due to the functional redundancy between Kþ
efflux mechanisms driving stomatal closure (see last entry of
Table 1), and the stabilization of the other regulators of the
node ‘‘Closure,’’ a closed steady state (Closure ¼ 1) is attained
within eight steps for any initial condition. The details of this
analysis will be published elsewhere.
Identification of Essential Components
After testing the wild-type (intact) system, we investigate
whether the disruption (loss) of a component changes the
system’s response to ABA. We systematically perturb the
system by setting the state of a node to 0 (off state), and
holding it at 0 for the duration of the simulation. This
perturbation mimics the effect of a knockout mutation for a
gene or pharmaceutical inhibition of secondary messenger
production or of kinase or phosphatase activity. We
characterize the effect of the node disruption by calculating
the percentage (probability) of closure response to a constant
ABA signal at each time step and comparing it with the
percentage of closure in the wild-type system.
The perturbed system’s responses can be classified into five
categories with respect to the system’s steady state and the
time it takes to reach the steady state. We designate responses
identical or very close to the wild-type response as having
normal sensitivity; in these cases the probability of closure
reaches 100% within eight timesteps. Disruptions that cause
the percentage of closed stomata to decrease to zero after the
first few steps are denoted as conferring ABA insensitivity (in
accord with experimental nomenclature). We observe re-
sponses where the probability of closure (the percentage of
stomata closed at any given timestep) settles at a nonzero
value that is less than 100%; we classify these responses as
having reduced sensitivity. Finally, in two classes of behavior
the probability of closure ultimately reaches 100%, but with a
different timing than the normal response. We refer to a
response with ABA-induced closure that is slower than wild-
type as hyposensitivity, while hypersensitivity corresponds to
ABA-induced closure that is faster than wild-type. Therefore,
the perturbed system’s responses can be classified into five
categories in the order of decreasing sensitivity defect:
insensitivity to ABA, reduced sensitivity, hyposensitivity,
normal sensitivity, and hypersensitivity.
We find that 25 single node disruptions (65%; compare
with Table 2) do not lead to qualitative effects: 100% of the
population responds to ABA with timecourses very close to
the wild-type response. In contrast, the loss of membrane
depolarizability, the disruption of anion efflux, and the loss of
actin cytoskeleton reorganization present clear vulnerabil-
ities: irrespective of initial conditions or of relative timing, all
simulated stomata become insensitive to ABA (Figure 5A).
Indeed, membrane depolarization is a necessary condition of
Kþ efflux, which is a necessary condition of closure, as is actin
cytoskeleton reorganization and anion efflux. The individual
disruption of seven other components—PLD, PA, SphK, S1P,
GPA1, Kþ efflux through slowly activating Kþ channels at the
plasma membrane (KOUT), and pHc increase —reduces ABA
sensitivity, as the percentage of closed stomata in the
population decreases to 20%—80% (see Figure 5B). At least
five components (S1P, SphK, PLD, PA, pHc) of these 7
predicted components have been shown to impair ABA-
Figure 5. The Probability of ABA-Induced Closure (i.e., the Percentage of
Simulations that Attain Closure) as a Function of Timesteps in the
Dynamic Model
In all panels, black triangles with dashed lines represent the normal (wild-
type) response to ABA stimulus. Open triangles with dashed lines show
that in wild-type, the probability of closure decays in the absence of ABA.
(A) Perturbations in depolarization (open diamonds) or anion efflux at
the plasma membrane (open squares) cause total loss of ABA-induced
closure. The effect of disrupting actin reorganization (not shown) is
identical to the effect of blocking anion efflux.
(B) Perturbations in S1P (dashed squares), PA (dashed circles), or pHc
(dashed diamonds) lead to reduced closure probability. The effect of
disrupting SphK is nearly identical to the effect of disrupting S1P (dashed
squares); perturbations in GPA1 and PLD, KOUT are very close to
perturbations in PA (dashed circles); for clarity, these curves are not
shown in the plot.
(C) abi1 recessive mutants (black squares) show faster than wild-type
ABA-induced closure (ABA hypersensitivity). The effect of blocking Ca2þ
ATPase(s) (not shown) is very similar to the effect of the abi1 mutation.
Blocking Ca2þ
c increase (black diamonds) causes slower than wild-type
ABA-induced closure (ABA hyposensitivity). The effect of disrupting
atrboh or ROS production (not shown) is very similar to the effect of
blocking Ca2þ
c increase.
DOI: 10.1371/journal.pbio.0040312.g005
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1739
Model of Guard Cell ABA Signaling
induced closure when clamped or mutated experimentally
[8,31,43,58]. For these disruptions, both theoretical analysis
and numerical results indicate that all simulated stomata
converge to limit cycles (oscillations) driven by the Ca2þ
c
oscillations, yet the ratio of open and closed stomata in the
population is the same at any timepoint, leading to a constant
probability of closure. (The alternative possibility, of a subset
of stomata being stably closed and another subset stably open,
was not observed for any disruption.)
For all other single-node disruptions the probability of
closure ultimately reaches 100% (i.e., all simulated stomata
reach the closed steady state); however, the rate of con-
vergence diverges from the rate of the wild-type response (see
Figure 5C). Disruption of Ca2þ
c increase or of the production
of ROS leads to ABA hyposensitivity (slower than wild-type
response). In contrast, the disruption of ABI1 or of the Ca2þ
ATPase(s) leads to ABA hypersensitivity (faster than wild
type-response) (Figure 5C). The hyposensitive and hyper-
sensitive responses are statistically distinguishable (p , 0.05
for all intermediary time steps [i.e., for 0 , t , 8]) from the
normal responses. Our model predicts that perturbation of
OST1 leads to a slower than normal response that is
nevertheless not slow enough to be classified as hyposensitive.
Indeed, ost1 mutants are still responsive to ABA even though
not as strongly as wild-type plants [12].
After analyzing all single knockout simulations, we turned
to analysis of double and triple knockout simulations. First, to
effectively distinguish between normal, hypo- and hyper-
sensitive responses (all of which achieve 100% probability of
closure, but at different rates), we calculated the cumulative
percentage of closure (CPC) by adding the probability of
closure over 12 steps; the smaller the CPC value, the more
slowly the probability of closure reaches 100%, and vice
versa. Plotting the histogram of CPC values reveals a clear
separation into three distinct groups of response in the case
of single disruptions (Figure 6A). In contrast, the cumulative
effects of multiple perturbations lead to a continuous
distribution of sensitivities in a broad range around the
normal (Figure 6B and 6C). We use the single perturbation
results to identify three classes of response that achieve 100%
closure, but at varying rates. We define two CPC thresholds:
the midpoint between the most hyposensitive single mutant
and normal response, CPChypo ¼ 10.35; and the midpoint
between the normal and least hypersensitive single mutant
response, CPChyper ¼ 10.7. Disruptions with cumulative
closure probability , CPChypo are classified as hyposensitive,
disruptions with cumulative closure probability . CPChyper
are hypersensitive; and values between the two thresholds are
classified as normal responses. This hypo/hypersensitive
classification does not affect the determination of insensitive
or reduced sensitivity responses, which are identified by
observing a null or less than 100% probability of closure.
For double (triple) knockout simulations, some combina-
tions of perturbations exhibit sensitivities that are independ-
ent of the sensitivity of each of their components’
perturbation. Normal ABA-induced stomatal closure is
Table 2. Single to Triple Node Disruptions in the Dynamic Model
Number of
Nodes Disrupted
Percentage with
Normal Sensitivity
Percentage
Causing Insensitivity
Percentage Causing
Reduced Sensitivity
Percentage Causing
Hyposensitivity
Percentage Causing
Hypersensitivity
1
65%
7.5%
17.5%
5%
5%
2
38%
16%
27%
12%
6%
3
23%
25%
31%
13%
7%
In all the perturbations, there are five groups of responses. Normal sensitivity refers to a response close to the wild-type response (shown as black triangles and dashed line in Figure 5).
Insensitivity means that the probability of closure is zero after the first three steps (see Figure 5A). Reduced sensitivity means that the probability of closure is less than 100% (see dashed
symbols in Figure 5B). Hyposensitivity corresponds to ABA-induced closure that is slower than wild-type (black diamonds in Figure 5C). Hypersensitivity corresponds to ABA-induced
closure that is faster than wild-type (black squares in Figure 5C).
DOI: 10.1371/journal.pbio.0040312.t002
Figure 6. Classification of Close-to-Normal Responses
(A) For all the single mutants that ultimately reach 100% closure, we plot
the histogram of the cumulative probability of closure (CPC). We find
three distinct types of responses: hypersensitivity (CPC . 10.7, for abi1
and Ca2þ ATPase disruption); hyposensitivity (CPC , 10.35, for Ca2þ
c ,
atrboh, and ROS disruption); and normal responses ( 10.35 , CPC ,
10.7). For all the double (B) and triple (C) mutants that eventually reach
100% closure at steady state when ABA ¼ 1, we classify the responses
using the CPC thresholds defined by the single mutant responses. The
CPC threshold values are indicated by dashed vertical lines in the plot.
DOI: 10.1371/journal.pbio.0040312.g006
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1740
Model of Guard Cell ABA Signaling
preserved in 38% (23%) of combinations (see Table 2). In
contrast, ABA signaling is completely blocked in 16% (25%)
of disruptions. In addition to perturbations involving the
three previously found insensitivity-causing single knockouts
(loss of membrane depolarizability, the disruption of anion
efflux, and the loss of actin cytoskeleton reorganization), a
large number of novel combinations are found. Interestingly,
perturbations of Ca2þ
c or Ca2þ release from stores, when
combined with disruptions in PLD, PA, GPA1, or pHc, lead to
insensitivity (see Figure 7 and Discussion). ABA-induced
closure is reduced (but not lost entirely) in 27% (31%) of the
cases. Hyposensitive responses are found for 12% (13%) of
double (triple) perturbations. All of the double perturbations
in this category involve a knockout mutation of Ca2þ
c,
Atrboh, or ROS. The triple perturbations involve a knockout
mutation of Ca2þ
c, Atrboh, or ROS, plus two other perturba-
tions, or combinations of three disruptions that alone are not
predicted to cause quantifiable effects (e.g., guanyl cyclase,
Ca2þ release from internal stores [CIS], and CaIM; see Figure
7). Around 6% (7%) of double (triple) perturbations, all
including a knockout mutation of ABI1 or Ca2þ ATPase, lead
to a hypersensitive response. In summary, accumulating
perturbations cause a dramatic decrease in the percentage
of normal response; the majority of triple knockouts are
either insensitive or have reduced sensitivity. The fraction of
hyposensitive and hypersensitive knockouts increases only
moderately.
Experimental Assessment of Model Predictions
As a first step toward experimental assessment of the
model’s predictions, we used a weak acid, Na-butyrate, to
clamp cytosolic pH, and then we treated the stomata with 50
lM ABA and observed the stomatal aperture responses. As
shown in Figure 8A, the stomatal aperture distributions
without butyrate treatments shift towards smaller apertures
after ABA treatment, forming a distribution that overlaps
with, but is clearly distinguishable from, the 0 ABA
distribution. However, when increasing concentrations of
butyrate are added in the solution, the ‘‘open’’ (0 ABA) and
‘‘closed’’ (þ ABA) distributions become increasingly over-
lapping (Figure 8B–8D). At the highest butyrate concentra-
tion (5 mM; Figure 8D), the 0 ABA and þABA populations of
stomatal apertures are statistically identical (the null hypoth-
esis that the two distributions are the same cannot be
Figure 7. Summary of the Dynamic Effects of Calcium Disruptions
All curves represent the probability of ABA-induced closure (i.e., the
percentage of simulations that attain closure) as a function of time steps.
Black triangles with dashed line represent the normal (wild-type)
response to ABA stimulus; open triangles with dashed lines show how
the probability of closure decays in the absence of ABA. CIS þ PA double
mutants (dashed circles) and Ca2þ
c þ pHc double mutants (dashed
diamonds) show insensitivity to ABA. Ca2þ ATPase þ RCN1 double
mutants (black circles) show hyposensitive (delayed) response to ABA.
Guanyl cyclase þ CIS þ CaIM triple mutants (black diamonds) also show
hyposensitivity; note that none of the guanyl cyclase or CIS or CaIM
single knockouts show changed sensitivity (data not shown). Ca2þ
ATPase mutants (black squares) show faster than wild-type ABA-induced
closure (ABA hypersensitivity).
DOI: 10.1371/journal.pbio.0040312.g007
Figure 8. Effect of Cytosolic pH Clamp (Increasing Concentrations of Na-
butyrate from 0 to 5 mM) on ABA-Induced Stomatal Closure
The histograms show the distribution of stomatal apertures without ABA
treatment (gray bars) and with 50 lM ABA (white bars). Throughout, the
x-axis gives the stomatal aperture size and the y-axis indicates the
fraction of stomata for which that aperture size was observed. The black
columns indicate the overlap between the 0 lM ABA and the 50 lM ABA
distributions. Note that the data of (A) and those of Figure 3A are
identical; these data are reproduced here for ease of comparison with
panels (B–D).
DOI: 10.1371/journal.pbio.0040312.g008
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1741
Model of Guard Cell ABA Signaling
rejected; two-tailed t test, p . 0.05). These results qualitatively
support our prediction of the importance of pHc signaling.
For a more quantitative comparison with the theoretically
predicted probability of closure corresponding to pH
clamping, one can define a threshold C between open and
closed stomatal states, such that stomata with apertures larger
than C can be classified as open and stomata with lower
apertures can be classified as closed. We identify the thresh-
old value C ¼ 4.3 lm by simultaneously minimizing the
fraction of stomata classified as closed in the control
condition and maximizing this fraction in the ABA treated
condition. Using this threshold we find that the fraction of
closed stomata in the 50 lM ABA þ 5 mM Na-butyrate
population is 26%, in agreement with the theoretically
predicted probability of closure (Figure 5B).
In plant systems, cytosolic pH changes in response to
multiple hormones such as ABA [20,59], jasmonates [21],
auxin [59], etc. The downstream effectors of pH changes
include ion channels [8], protein kinases [60], and protein
phosphatases [30]. Previous experiments with guard cells have
demonstrated the efficacy of butyrate in imposing a cytosolic
pH clamp [8,21]. While these prior experiments focused on a
single concentration of butyrate, here we used five different
concentrations (three shown), with 120 stomata sampled for
each treatment. As seen in Figure 8, we were able to monitor
the effect of butyrate in the þABA treatment in both
increasing the mean aperture size and reducing the spread
of the aperture sizes. There is a clear indication of saturation
between the two highest butyrate concentrations. While
detailed measurements of cytosolic pH constitute a full
separate study beyond the scope of the present article, the
results of Figure 8 support the suggestion from our model
that pHc should receive increased attention by experimen-
talists as a focal point for transduction of the ABA signal.
Discussion
Network Synthesis and Path Analysis
Logical organization of large-scale data sets is an important
challenge in systems biology; our model provides such
organization for one guard cell signaling system. As summar-
ized in Table S1, we have organized and formalized the large
amount of information that has been gathered on ABA
induction of stomatal closure from individual experiments.
This information has been used to reconstruct the ABA
signaling network (Figure 2). Figure 2 uses different types of
edges (lines) to depict activation and inhibition, and also uses
different edge colors to indicate whether the information was
derived from our model species, Arabidopsis, or from another
plant species. Different types of nodes (metabolic enzymes,
signaling proteins, transporters, and small molecules) are also
color coded. An advantage of our method of network
construction over other methods such as those used in
Science’s Signal Transduction Knowledge Environment
(STKE) connection maps [61] is the inclusion of intermediate
nodes when direct physical interactions between two compo-
nents have not been demonstrated.
As is evident from Figure 2, network synthesis organizes
complex information sets in a form such that the collective
components and their relationships are readily accessible.
From such analysis, new relationships are implied and new
predictions can be made that would be difficult to derive
from less formal analysis. For example, building the network
allows one to ‘‘see’’ inferred edges that are not evident from
the disparate literature reports. One example is the path
from S1P to ABI1 through PLD. Separate literature reports
indicate that PLDa null mutants show increased transpira-
tion, that PLDa1 physically interacts with GPA1, that S1P
promotion of stomatal closure is reduced in gpa1 mutants,
that PLD catalyses the production of PA, and that recessive
abi1 mutants are hypersensitive to ABA. Network inference
allows one to represent all this information as the S1P !
GPA1 ! PLD ! PA—j ABI1—j closure path, and make the
prediction that ABA inhibition of ABI1 phosphatase activity
will be impaired in sphingosine kinase mutants unable to
produce S1P.
Another prediction that can be derived from our network
analysis is a remarkable redundancy of ABA signaling, as
there are eight paths that emanate from ABA in Figure 2 and,
based on current knowledge (though see below) these paths
are initially independent. The prediction of redundancy is
consistent with previous, less formal analyses [62]. The
integrated guard cell signal transduction network (which
includes the ABA signal transduction network) has been
proposed as an example of a robust scale-free network [62].
To classify a network as scale-free, one needs to determine
the degree (the number of edges, representing interactions/
regulatory relationships) of each node, and to calculate the
distribution of node degrees (denoted degree distribution)
[45,46]. Scale-free networks, characterized by a degree
distribution described by a power law, retain their connec-
tivity in the face of random node disruptions, but break down
when the highest-degree nodes (the so-called hubs) are lost
[46]. While the guard cell network may ultimately prove to be
scale-free, the network is not sufficiently large at present to
verify the existence of a power-law degree distribution; thus,
the analogy with scale-free networks cannot be rigorously
satisfied.
Dynamic Modeling
Our model differs from previous models employed in the
life sciences in the following fundamental aspects. First, we
have reconstructed the signaling network from inferred
indirect relationships and pathways as opposed to direct
interactions; in graph theoretical terminology, we found the
minimal network consistent with a set of reachability
relationships. This network predicts the existence of numer-
ous additional signal mediators (intermediary nodes), all of
which could be targets of regulation. Second, the network
obtained is significantly more complex than those usually
modeled in a dynamic fashion. We bridge the incompleteness
of regulatory knowledge and the absence of quantitative
dose-response relationships for the vast majority of the
interactions in the network by employing qualitative and
stochastic dynamic modeling previously applied only in the
context of gene regulatory networks [53].
Mathematical models of stomatal behavior in response to
environmental change have been studied for decades [63,64].
However, no mathematical model has been formulated that
integrates the multitude of recent experimental findings
concerning the molecular signaling network of guard cells.
Boolean modeling has been used to describe aspects of plant
development such as specification of floral organs [65], and
there are a handful of reports describing Boolean models of
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1742
Model of Guard Cell ABA Signaling
light and pathogen-, and light by carbon-regulated gene
expression [66–68]. Use of a qualitative modeling framework
for signaling networks is justified by the observation that
signaling networks maintain their function even when faced
with fluctuations in components and reaction rates [69]. Our
model uses experimental evidence concerning the effects of
gene knockouts and pharmacological interventions for
inferring the downstream targets of the corresponding gene
products and the sign of the regulatory effect on these
targets. However, use of this information does not guarantee
that the dynamic model will reproduce the dynamic outcome
of the knockout or intervention. Indeed, all model ingre-
dients (node states, transfer functions) refer to the node
(component) level, and there is no explicit control over
pathway-level effects. Moreover, the combinatorial transfer
functions we employed are, to varying extents, conjectures,
informed by the best available experimental information (see
Text S1). Finally, in the absence of detailed knowledge of the
timing of each process and of the baseline (resting) activity of
each component, we deliberately sample timescales and
initial conditions randomly. Thus, an agreement between
experimental and theoretical results of node disruptions is
not inherent, and would provide a validation of the model.
The accuracy of our model is indeed supported by its
congruency with experimental observation at multiple levels.
At the pathway level, our model captures, for example, the
inhibition of ABA-induced ROS production in both ost1
mutants and atrboh mutants [12,19,21] and the block of ABA-
induced stomatal closure in a dominant-positive atRAC1
mutant [22]. In our model, as in experiments, ABA-induced
NO production is abolished in either nos single or nia12
double mutants [13,18]. Moreover, the model reproduces the
outcome that ABA can induce cytosolic Kþ decrease by Kþ
efflux through the alternative potassium channel KAP, even
when ABA-induced NO production leads to the inhibition of
the outwardly-rectifying (KOUT) channel [70]. At the level of
whole stomatal physiology, our model captures the findings
that anion efflux [35,71] and actin cytoskeleton reorganiza-
tion [22] are essential to ABA-induced stomatal closure. The
importance of other components such as PA, PLD, S1P,
GPA1,
KOUT,
pH c
in
stomatal
closure
control
[8,20,31,43,58,72], and the ABA hypersensitivity conferred
by elimination of signaling through ABI1 [28,29], are also
reproduced. Our model is also consistent with the observa-
tion that transgenic plants with low PLC expression still
display ABA sensitivity [73].
The fact that our model accords well with experimental
results suggests that the inferences and assumptions made are
correct overall, and enables us to use the model to make
predictions about situations that have yet to be put to
experimental test. For example, the model predicts that
disruption of all Ca2þ ATPases will cause increased ABA
sensitivity, a phenomenon difficult to address experimentally
due to the large family of calcium ATPases expressed in
Arabidopsis guard cells (unpublished data). Most of the
multiple perturbation results presented in Figure 5 and
Table 2 also represent predictions, as very few of them have
been tested experimentally. Results from our model can now
be used by experimentalists to prioritize which of the
multitude of possible double and triple knockout combina-
tions should be studied first in wet bench experiments.
Most importantly, our model makes novel predictions
concerning the relative importance of certain regulatory
elements. We predict three essential components whose
elimination completely blocks ABA-induced stomatal closure:
membrane depolarization, anion efflux, and actin cytoskele-
ton reorganization. Seven components are predicted to
dramatically affect the extent and stability of ABA-induced
stomatal closure: pHc control, PLD, PA, SphK, S1P, G protein
signaling (GPA1), and Kþ efflux. Five additional components,
namely increase of cytosolic Ca2þ, Atrboh, ROS, the Ca2þ
ATPase(s), and ABI1, are predicted to affect the speed of
ABA-induced stomatal closure. Note that a change in
stomatal response rate may have significant repercussions,
as some stimuli to which guard cells respond fluctuate on the
order of seconds [74,75]. Thus our model predicts two
qualitatively different realizations of a partial response to
ABA: fluctuations in individual responses (leading to a
reduced steady-state sensitivity at the population level), and
delayed response. These predictions provide targets on which
further experimental analysis should focus.
Six of the 13 key positive regulators, namely increase of
cytosolic Ca2þ, depolarization, elevation of pHc, ROS, anion
efflux, and Kþ efflux through outwardly rectifying Kþ
channels, can be considered as network hubs [45], as they
are in the set of ten highest degree (most interactive) nodes.
Other nodes whose disruption leads to reduced ABA
sensitivity, namely SphK, S1P, GPA1, PLD, and PA, are part
of the ABA ! PA path. While they are not highly connected
themselves, their disruption leads to upregulation of the
inhibitor ABI1, thus decreasing the efficiency of ABA-
induced stomatal closure. Similarly, the node representing
actin reorganization has a low degree. Thus the intuitive
prediction, suggested by studies in yeast gene knockouts
[76,77], that there would be a consistent positive correlation
between a node’s degree and its dynamic importance, is not
supported here, providing another example of how dynamic
modeling can reveal insights difficult to achieve by less formal
methods. This lack of correlation has also been found in the
context of other complex networks [78].
Comparing Figure 3 and Figure 6C, one can notice a
similar heterogeneity in the measured stomatal aperture size
distributions and the theoretical distribution of the cumu-
lative probability of closure in the case of multiple node
disruptions. While apparently unconnected, there is a link
between the two types of heterogeneity. Due to stochastic
effects on gene and protein expression, it is possible that in a
real environment not all components of the ABA signal
transduction network are fully functional. Therefore, even
genetically identical populations of guard cells may be
heterogeneous at the regulatory and functional level, and
may respond to ABA in slightly different ways. In this case,
the heterogeneity in double and triple disruption simulations
provides an explanation for the observed heterogeneity in
the experimentally normal response: the latter is actually a
mixture of responses from genetically highly similar but
functionally nonidentical guard cells.
Importance of Ca2þ
c Oscillations to ABA-Induced
Stomatal Closure
Through the inclusion of the nodes CaIM, CIS, and the
Ca2þ ATPase node representing the Ca2þ ATPases and Ca2þ/
Hþ antiporters [79,80] that drive Ca2þ efflux from the
cytosolic compartment, our model incorporates the phenom-
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1743
Model of Guard Cell ABA Signaling
enon of oscillations in cytosolic Ca2þ concentration, which
has been frequently observed in experimental studies
[56,81,82]. In experiments where Ca2þ
c is manipulated,
imposed Ca2þ
c oscillations with a long periodicity (e.g., 10
min of Ca2þ elevation with a periodicity of once every 20 min)
are effective in triggering and maintaining stomatal closure,
yet at 10 min (i.e., after just one Ca2þ
c transient elevation and
thus before the periodicity of the Ca2þ change can be
‘‘known’’ by the cell), significant stomatal closure has already
occurred [56]. This result suggests that the Ca2þ
c oscillation
signature may be more important for the maintenance of
closure than for the induction of closure [56,81], and that the
induction of closure might only be dependent on the first,
transient Ca2þ
c elevation.
According to our model, if Ca2þ
c elevation occurs, then
stomatal closure is triggered (consistent with numerous
experimental studies), but Ca2þ
c elevation is not required
for ABA-induced stomatal closure. Re-evaluation of the
experimental studies on ABA and Ca2þ
c reveals support for
this prediction. First, although Ca2þ elevation certainly can be
observed in guard cell responses to ABA, numerous exper-
imental results also show that Ca2þ
c elevation is only observed
in a fraction of the guard cells assayed [9,83]. Furthermore,
absence of Ca2þ
c elevation in response to ABA does not
prevent the occurrence of downstream events such as ion
channel regulation [84,85] and stomatal closure [86,87], a
phenomenon also predicted by our in silico analysis. Second,
it has been observed that some guard cells exhibit sponta-
neous oscillations in Ca2þ
c, and in such cells, ABA application
actually suppresses further Ca2þ
c elevation [88]; thus, ABA
and Ca2þ
c elevation are clearly decoupled.
Our model does predict that disruption of Ca2þ signaling
leads to ABA hyposensitivity, or a slower than normal
response to ABA. In the real-world environment, even a
slight delay or change in responsiveness may have significant
repercussions, as some stimuli to which guard cells respond
fluctuate on the order of seconds; and stomatal responses can
have comparable rapidity [74,75]. Moreover, our model
predicts that Ca2þ
c elevation (although not necessarily
oscillation) becomes required for engendering stomatal
closure when pHc changes, Kþ efflux or the S1P–PA pathway
are perturbed (see Figure 7). Thus, Ca2þ
c modulation confers
an essential redundancy to the network. Support for such a
redundant role can be found in a study by Webb et al. [89]
where Ca2þ concentration was reduced below normal resting
levels by intracellular application of BAPTA (such reduction
in baseline Ca2þ
c levels has been shown to reduce ABA
activation of anion channels [85]) and the epidermal tissue
was perfused with CO2-free air, a treatment that has been
shown to inhibit outwardly rectifying Kþ channels and slow
anion efflux channels [90]. The ABA insensitivity of stomatal
closure found by Webb et al. under these conditions [89]
therefore can be attributed to a combination of multiple
perturbations (of Ca2þ
c elevation, Kþ efflux, and anion efflux)
and is consistent with the predictions of our model.
Our model indicates that double perturbations of the Ca2þ
ATPase component and either of RCN1, OST1, NO, NOS,
NIA12, or Atrboh are hyposensitive (see Figure 7), consistent
with experimental results on disruptions in the latter
components [12,13,18,19,21,91]. Since the latter disruptions
alone, with unperturbed Ca2þ ATPase, are found to have a
close-to-normal response in our model, a Ca2þ ATPase–
disrupted and therefore Ca2þ
c oscillation–free model seems
to be closer to experimental observations on stomatal
aperture response recorded for these individual mutant
genotypes. This suggests that Ca2þ
c elevation (and not Ca2þ
c
oscillation) is the signal perceived by downstream factors that
control the induction of closure. Possibly, certain as-yet-
undiscovered interaction motifs, such as a synergistic feed-
forward loop [92] or dual positive feedback loops [93], could
transform the Ca2þ
c oscillation into a stable downstream
output.
Limitations of the Current Analysis
Network topology. Our graph reconstruction is incom-
plete, as new signaling molecules will certainly be discovered.
Novel nodes may give identity to the intermediary nodes that
our model currently incorporates. Discovery of a new
interaction among known nodes could simplify the graph
by reducing (apparent) redundancy. For example, if it is
found that GPA1 ! OST1, the simplest interpretation of the
ABA ! ROS pathway becomes ABA ! GPA1 ! OST1 !
ROS, and the graph loses one edge and an alternative
pathway. As an effect, the graph’s robustness will be
attenuated. Among likely candidates for network reduction
are the components currently situated immediately down-
stream of ABA because, in the absence of information about
guard cell ABA receptors [94], we assumed that ABA
independently regulates eight components. It is also possible
that a newly found interaction will not change the existing
edges, but only add a new edge. A newly added positive
regulation edge will further increase the redundancy of
signaling and correspondingly its robustness. Newly added
inhibitory edges could possibly damage the network’s robust-
ness if they affect the main positive regulators of the network,
especially anion channels and membrane depolarization. For
example, experimental evidence indicates that abi1 abi2
double recessive mutants are more sensitive to ABA-induced
stomatal closure than abi1 or abi2 single recessive mutants
[29], suggesting that ABI1 and ABI2 act synergistically. Due to
limited experimental evidence, we do not explicitly incorpo-
rate ABI2, but an independent inhibitory effect of ABI2
would diminish ABA signaling.
While it is difficult to estimate the changes in our
conclusions due to future knowledge gain, we can gauge the
robustness of our results by randomly deleting entries in
Table S1 or rewiring edges of Figure 2 (see Texts S2 and S3).
We find that most of the predicted important nodes are
documented in more than one entry, and more than one
entry needs to be removed from the database before the
topology of the network related to that node changes (Text
S2). Random rewiring of up to four edge pairs shows that the
dynamics of our current network is moderately resilient to
minor topology changes (Text S3 and Figure S1).
Dynamic model. In our dynamic model we do not place
restrictions on the relative timing of individual interactions
but sample all possible updates randomly. This approach
reflects our lack of knowledge concerning the relative
reaction speeds as well as possible environmental noise. The
significance of our current results is the prediction that
whatever the timing is, given the current topology of
regulatory relationships in the network, the most essential
regulators will not change. Our approach can be iteratively
refined when experimental results on the strength and timing
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1744
Model of Guard Cell ABA Signaling
of individual interactions become available. For example, we
can combine Boolean regulation with continuous synthesis
and degradation of small molecules or signal transduction
proteins [95,96] as kinetic (rate) data emerge. Our model
considers the response of individual guard cell pairs to the
local ABA signal; however, there is recent evidence of a
synchronized oscillatory behavior of stomatal apertures over
spatially extended patches in response to a decrease in
humidity [97]. Our model can be extended to incorporate
cell-to-cell signaling and spatial aspects by including extrac-
ellular regulators when information about them becomes
available (see [51]).
Node disruptions. A knockout may either deprive the
system of an essential signaling element (the gene itself), or it
may ‘‘set’’ the entire system into a different state (e.g., by
affecting the baseline expression of other, seemingly unre-
lated signaling elements). Our analysis and current exper-
imental data only address the former. Because of this caveat,
in some ways rapid pharmacological inhibition may actually
have a more specific effect on the cell than gene knockouts.
Implications
Many of the signaling proteins present as nodes in our
model are represented by multigene families in Arabidopsis
[98], with likely functional redundancy among encoded
isoforms. Therefore, the amount of experimental work
required to completely disrupt a given node may be
considerable. It is also considerable work to make such
genetic modification in many of the important crop species
that are much less amenable than Arabidopsis to genetic
manipulation. It is also the case that, at present, there are no
reports of successful use of ratiometric pH indicators in the
small guard cells of Arabidopsis, suggesting that further
technical advances in this area are required. Facts such as
these indicate the importance of establishing a prioritization
of node disruption in experimental studies seeking to
manipulate stomatal responses for either an increase in basic
knowledge or an improvement in crop water use efficiency.
Our model provides information on which such prioritiza-
tion can be based. Future work on this model will focus on
predicting the changes in ABA-induced closure upon con-
stitutive activation of network components or in the face of
fluctuating ABA signals. Ultimately, the experimental infor-
mation obtained may or may not support the model
predictions; the latter instance provides new information
that can be used to improve the model. Through such
iteration of in silico and wet bench approaches, a more
complete understanding of complex signaling cascades can
be obtained.
Approaches to describe the dynamics of biological net-
works include differential equations based on mass-action
kinetics for the production and decay of all components
[99,100], and stochastic models that address the deviations
from population homogeneity by transforming reaction rates
into probabilities and concentrations into numbers of
molecules [101]. The great complexity of many cellular signal
transduction networks makes it a daunting task to recon-
struct all the reactions and regulatory interactions in such
explicit biochemical and kinetic detail. Our work offers a
roadmap for synthesizing incompletely described signal
transduction and regulatory networks utilizing network
theory and qualitative stochastic dynamic modeling. In
addition to being the practical choice, qualitative dynamic
descriptions are well suited for networks that need to
function robustly despite changes in external and internal
parameters. Indeed, several analyses found that the dynamics
of network motifs crucial for the stable dynamics and noise-
resistance of cellular networks, such as single input modules,
feed-forward loops [102,103] and dual positive feedback loops
[93], is correctly and completely captured by qualitative
modeling [104,105]. For example, at the regulatory module
level, several qualitative (Boolean and continuous/discrete
hybrid) models [51,53,96] reproduced the Drosophila segment
polarity gene network’s resilience when facing variations in
kinetic parameters [50], offering the most natural explan-
ation of which parameter sets will succeed in forming the
correct gene expression pattern [106]. We expect that our
methods will find extensive applications in systems where
modeling is currently not possible by traditional approaches
and that they will act as a scaffold on which more quantitative
analyses of guard cell signaling in particular and cell signaling
in general can later be built.
Our analyses have clear implications for the design of future
wet bench experiments investigating the signaling network of
guard cells and for the translation of experimental results on
model species such as Arabidopsis to the improvement of water
use efficiency and drought tolerance in crop species [107–
109]. Drought stress currently provides one of the greatest
limitations to crop productivity worldwide [110,111], and this
issue is of even more concern given current trends in global
climate change [112,113]. Our methods also have implications
in biomedical sciences. The use of systems modeling tools in
designing new drugs that overcome the limitation of tradi-
tional medicine has been suggested in the recent literature
[114]. Many human diseases, such as breast cancer [115] or
acute myeloid leukemia [116,117], cause complex alterations
to the underlying signal transduction networks. Pathway
information relevant to human disease etiologies has been
accumulated over decades and such information is stored in
several databases such as TRANSPATH [118], BioCarta (http://
www.biocarta.com), and STKE (http://www.stke.org). Our
strategy can serve as a tool that guides experiments by
integrating qualitative data, building systems models, and
identifying potential drug targets.
Materials and Methods
Plant material and growth conditions. Wild-type Arabidopsis (Col
genotype) seeds were germinated on 0.53MS media plates containing
1% sucrose. Seedlings were grown vertically under short-day
conditions (8 h light/16 h dark) 120 lmol m2 s1 for 10 d. Vigorous
seedlings were selected for transplantation into soil and were grown
to 5 wk of age (from germination) under short day conditions (8 h
light/16 h dark). Leaves were harvested 30 min after the lights were
turned on in the growth chamber.
Stomatal aperture measurements. Leaves were incubated in 20 mM
KCl, 5 mM Mes-KOH, and 1 mM CaCl2 (pH 6.15) (Tris), at room
temperature and kept in the light (250 lmol m2 s1) for 2 h to open
stomata. For pHc clamping, different amounts of Na-butyrate stock
solution (made up as 1M solution in water [pH 6.1]) were added into
the incubation solution, to achieve the concentrations given in Figure
8, 15 min before adding 50 lM ABA. Apertures were recorded after
2.5 h of further incubation in light. Epidermal peels were prepared at
the end of each treatment. The maximum width of each stomatal
pore was measured under a microscope fitted with an ocular
micrometer. Data were collected from 40 stomata for each treatment
and each experiment was repeated three times.
Model. The network in Figure 2 was drawn with the SmartDraw
software (http://www.smartdraw.com/exp/ste/home). The dynamic
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1745
Model of Guard Cell ABA Signaling
modeling was implemented by custom Python code (http://www.
python.org). To equally sample the space of all possible timescales,
the random-order asynchronous updating method developed in [53]
was used. Briefly, every node is updated exactly once during each unit
time interval, according to a given order. This order is a permutation
of the N¼40 nodes in the network, chosen randomly out of a uniform
distribution over the set of all N! possible permutations. A new
update order is selected at each timestep. As demonstrated in [53],
this algorithm is equivalent to a random timing of each node’s state
transition.
Supporting Information
Figure S1. Probability of Closure in Randomized Networks where
Pairs of Positive or Negative Edges Are Rewired
Found at DOI: 10.1371/journal.pbio.0040312.sg001 (40 KB PDF).
Table S1. Synthesis of Experimental Information about Regulatory
Interactions between ABA Signal Transduction Pathway Components
Found at DOI: 10.1371/journal.pbio.0040312.st001 (407 KB DOC).
Text S1. Detailed Justification for Each Boolean Transfer Function
Found at DOI: 10.1371/journal.pbio.0040312.sd001 (149 KB DOC).
Text S2.
Verification of the Inference Process and the Resulting
Network
Found at DOI: 10.1371/journal.pbio.0040312.sd002 (45 KB DOC).
Text S3. Effect of Random Rewiring on the Network Dynamics
Found at DOI: 10.1371/journal.pbio.0040312.sd003 (36 KB DOC).
Accession Numbers
The Arabidopsis Information Resource (TAIR) (http://www.arabidopsis.
org) accession numbers for the genes discussed in this paper are
NIA12 (At1g77760/At1g37130), GPA1 (At2g26300), ERA1 (At5g40280),
AtrbohD/F (At5g47910/At4g11230), RCN1 (At1g25490), OST1
(At4g33950), ROP2 (At1g20090), RAC1 (At4g35020), ROP10
(At3g48040), AtP2C-HA/AtPP2CA (At1g72770/At3g11410), and GCR1
(At1g48270).
Acknowledgments
The authors thank Drs. Jayanth Banavar, Vincent Crespi, and Eric
Harvill for critically reading a previous version of the manuscript;
and Dr. Istva´n Albert for assistance with figure preparation.
Author contributions. SL, SMA, and RA conceived and designed
the experiments. SL performed the experiments. SL and RA analyzed
the data. SL, SMA, and RA wrote the paper.
Funding. RA gratefully acknowledges a Sloan Research Fellowship.
Research on guard cell signaling in SMA’s laboratory is supported by
NSF-MCB02–09694 and NSF-MCB03–45251.
Competing interests. The authors have declared that no competing
interests exist.
References
1.
Fall CP, Marland ES, Wagner JM, Tyson JJ (2002) Computational cell
biology. New York: Springer. 468 p.
2.
Voit EO (2000) Computational analysis of biochemical systems. Cam-
bridge: Cambridge University Press. 531 p.
3.
Bower JM, Bolouri, H. (2001) Computational modeling of genetic and
biochemical networks. Cambridge (Massachusetts): MIT Press. 336 p.
4.
Uetz P, Giot L, Cagney G, Mansfield TA, Judson RS, et al. (2000) A
comprehensive analysis of protein-protein interactions in Saccharomyces
cerevisiae. Nature 403: 623–627.
5.
Li S, Armstrong CM, Bertin N, Ge H, Milstein S, et al. (2004) A map of the
interactome network of the metazoan C. elegans. Science 303: 540–543.
6.
Schroeder JI, Allen GJ, Hugouvieux V, Kwak JM, Waner D (2001) Guard
cell signal transduction. Annu Rev Plant Physiol Plant Mol Biol 52: 627–
658.
7.
Peiter E, Maathuis FJ, Mills LN, Knight H, Pelloux J, et al. (2005) The
vacuolar Ca2þ-activated channel TPC1 regulates germination and stoma-
tal movement. Nature 434: 404–408.
8.
Wang XQ, Ullah H, Jones AM, Assmann SM (2001) G protein regulation of
ion channels and abscisic acid signaling in Arabidopsis guard cells. Science
292: 2070–2072.
9.
Blatt MR, Grabov A (1997) Signal redundancy, gates and integration in the
control of ion channels for stomatal movement. J Exp Bot 48: 529–537.
10.
Miedema H, Assmann SM (1996) A membrane-delimited effect of internal
pH on the Kþ outward rectifier of Vicia faba guard cells. J Membr Biol 154:
227–237.
11.
Pandey S, Assmann SM (2004) The Arabidopsis putative G protein-coupled
receptor GCR1 interacts with the G protein a subunit GPA1 and regulates
abscisic acid signaling. Plant Cell 16: 1616–1632.
12.
Mustilli AC, Merlot S, Vavasseur A, Fenzi F, Giraudat J (2002) Arabidopsis
OST1 protein kinase mediates the regulation of stomatal aperture by
abscisic acid and acts upstream of reactive oxygen species production.
Plant Cell 14: 3089–3099.
13.
Desikan R, Griffiths R, Hancock J, Neill S (2002) A new role for an old
enzyme: Nitrate reductase-mediated nitric oxide generation is required
for abscisic acid-induced stomatal closure in Arabidopsis thaliana. Proc Natl
Acad Sci U S A 99: 16314–16318.
14.
Aho A, Garey MR, Ullman JD. (1972) The transitive reduction of a
directed graph. SIAM J Comp 1: 131–137.
15.
Hunt L, Mills LN, Pical C, Leckie CP, Aitken FL, et al. (2003)
Phospholipase C is required for the control of stomatal aperture by
ABA. Plant J 34: 47–55.
16.
Lemtiri-Chlieh F, MacRobbie EA, Webb AA, Manison NF, Brownlee C, et
al. (2003) Inositol hexakisphosphate mobilizes an endomembrane store of
calcium in guard cells. Proc Natl Acad Sci U S A 100: 10091–10095.
17.
Garcia-Mata C, Gay R, Sokolovski S, Hills A, Lamattina L, et al. (2003)
Nitric oxide regulates Kþ and Cl channels in guard cells through a subset
of abscisic acid-evoked signaling pathways. Proc Natl Acad Sci U S A 100:
11116–11121.
18.
Guo FQ, Okamoto M, Crawford NM (2003) Identification of a plant nitric
oxide synthase gene involved in hormonal signaling. Science 302: 100–103.
19.
Kwak JM, Mori IC, Pei ZM, Leonhardt N, Torres MA, et al. (2003) NADPH
oxidase AtrbohD and AtrbohF genes function in ROS-dependent ABA
signaling in Arabidopsis. EMBO J 22: 2623–2633.
20.
Irving HR, Gehring CA, Parish RW (1992) Changes in cytosolic pH and
calcium of guard cells precede stomatal movements. Proc Natl Acad Sci U
S A 89: 1790–1794.
21.
Suhita D, Raghavendra AS, Kwak JM, Vavasseur A (2004) Cytoplasmic
alkalization precedes reactive oxygen species production during methyl
jasmonate- and abscisic acid-induced stomatal closure. Plant Physiol 134:
1536–1545.
22.
Lemichez E, Wu Y, Sanchez JP, Mettouchi A, Mathur J, et al. (2001)
Inactivation of AtRac1 by abscisic acid is essential for stomatal closure.
Genes Dev 15: 1808–1816.
23.
Hwang JU, Lee Y (2001) Abscisic acid-induced actin reorganization in
guard cells of dayflower is mediated by cytosolic calcium levels and by
protein kinase and protein phosphatase activities. Plant Physiol 125:
2120–2128.
24.
Du Z, Aghoram K, Outlaw WH Jr. (1997) In vivo phosphorylation of
phosphoenolpyruvate carboxylase in guard cells of Vicia faba L. is enhanced
by fusicoccin and suppressed by abscisic acid. Arch Biochem Biophys 337:
345–350.
25.
Dittrich P, Raschke K (1977) Malate metabolism in isolated epidermis of
Commelina communis L. in relation to stomatal functioning. Planta 134: 77–
81.
26.
Talbott LD, Zeiger E (1993) Sugar and organic acid accumulation in guard
cells of Vicia faba in response to red and blue light. Plant Physiol 102:
1163–1169.
27.
Talbott LD, Zeiger E (1996) Central roles for potassium and sucrose in
guard-cell osmoregulation. Plant Physiol 111: 1051–1057.
28.
Gosti F, Beaudoin N, Serizet C, Webb AA, Vartanian N, et al. (1999) ABI1
protein phosphatase 2C is a negative regulator of abscisic acid signaling.
Plant Cell 11: 1897–1910.
29.
Merlot S, Gosti F, Guerrier D, Vavasseur A, Giraudat J (2001) The ABI1
and ABI2 protein phosphatases 2C act in a negative feedback regulatory
loop of the abscisic acid signalling pathway. Plant J 25: 295–303.
30.
Leube MP, Grill E, Amrhein N (1998) ABI1 of Arabidopsis is a protein
serine/threonine phosphatase highly regulated by the proton and
magnesium ion concentration. FEBS Lett 424: 100–104.
31.
Zhang W, Qin C, Zhao J, Wang X (2004) Phospholipase Da1-derived
phosphatidic acid interacts with ABI1 phosphatase 2C and regulates
abscisic acid signaling. Proc Natl Acad Sci U S A 101: 9508–9513.
32.
Meinhard M, Grill E (2001) Hydrogen peroxide is a regulator of ABI1, a
protein phosphatase 2C from Arabidopsis. FEBS Lett 508: 443–446.
33.
Allen GJ, Kuchitsu K, Chu SP, Murata Y, Schroeder JI (1999) Arabidopsis
abi1–1 and abi2–1 phosphatase mutations reduce abscisic acid-induced
cytoplasmic calcium rises in guard cells. Plant Cell 11: 1785–1798.
34.
Pei ZM, Kuchitsu K, Ward JM, Schwarz M, Schroeder JI (1997) Differential
abscisic acid regulation of guard cell slow anion channels in Arabidopsis
wild-type and abi1 and abi2 mutants. Plant Cell 9: 409–423.
35.
Schwartz A, Ilan N, Schwarz M, Scheaffer J, Assmann SM, et al. (1995)
Anion-channel blockers inhibit S-type anion channels and abscisic acid
responses in guard cells. Plant Physiol 109: 651–658.
36.
Leonhardt N, Kwak JM, Robert N, Waner D, Leonhardt G, et al. (2004)
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1746
Model of Guard Cell ABA Signaling
Microarray expression analyses of Arabidopsis guard cells and isolation of a
recessive abscisic acid hypersensitive protein phosphatase 2C mutant.
Plant Cell 16: 596–615.
37.
Kuhn JM, Boisson-Dernier A, Dizon MB, Maktabi MH, Schroeder JI (2006)
The protein phosphatase AtPP2CA negatively regulates abscisic acid
signal transduction in Arabidopsis, and effects of abh1 on AtPP2CA mRNA.
Plant Physiol 140: 127–139.
38.
Hugouvieux V, Kwak JM, Schroeder JI (2001) An mRNA cap binding
protein, ABH1, modulates early abscisic acid signal transduction in
Arabidopsis. Cell 106: 477–487.
39.
Hugouvieux V, Murata Y, Young JJ, Kwak JM, Mackesy DZ, et al. (2002)
Localization, ion channel regulation, and genetic interactions during
abscisic acid signaling of the nuclear mRNA cap-binding protein, ABH1.
Plant Physiol 130: 1276–1287.
40.
Pei ZM, Ghassemian M, Kwak CM, McCourt P, Schroeder JI (1998) Role of
farnesyltransferase in ABA regulation of guard cell anion channels and
plant water loss. Science 282: 287–290.
41.
Allen GJ, Murata Y, Chu SP, Nafisi M, Schroeder JI (2002) Hypersensitivity
of abscisic acid-induced cytosolic calcium increases in the Arabidopsis
farnesyltransferase mutant era1–2. Plant Cell 14: 1649–1662.
42.
Kaiser WM, Weiner H, Kandlbinder A, Tsai CB, Rockel P, et al. (2002)
Modulation of nitrate reductase: Some new insights, an unusual case and a
potentially important side reaction. J Exp Bot 53: 875–882.
43.
Coursol S, Fan LM, Le Stunff H, Spiegel S, Gilroy S, et al. (2003)
Sphingolipid signalling in Arabidopsis guard cells involves heterotrimeric G
proteins. Nature 423: 651–654.
44.
Ma’ayan A, Jenkins SL, Neves S, Hasseldine A, Grace E, et al. (2005)
Formation of regulatory patterns during signal propagation in a
mammalian cellular network. Science 309: 1078–1083.
45.
Albert R (2005) Scale-free networks in cell biology. J Cell Sci 118: 4947–
4957.
46.
Albert R, Baraba´si AL (2002) Statistical mechanics of complex networks.
Rev Mod Physics 74: 47–49.
47.
Heimovaara-Dijkstra S, Heistek JC, Wang M (1994) Counteractive effects
of ABA and GA3 on extracellular and intracellular pH and malate in
barley aleurone. Plant Physiol 106: 359–365.
48.
Zhang SQ, Outlaw WH Jr., Chollet R (1994) Lessened malate inhibition of
guard-cell phosphoenolpyruvate carboxylase velocity during stomatal
opening. FEBS Lett 352: 45–48.
49.
Kauffman SA (1993) The origins of order: Self organization and selection
in evolution. New York: Oxford University Press. 709 p.
50.
von Dassow G, Meir E, Munro EM, Odell GM (2000) The segment polarity
network is a robust developmental module. Nature 406: 188–192.
51.
Albert R, Othmer HG (2003) The topology of the regulatory interactions
predicts the expression pattern of the segment polarity genes in Drosophila
melanogaster. J Theor Biol 223: 1–18.
52.
Thomas R (1973) Boolean formalization of genetic control circuits. J
Theor Biol 42: 563–585.
53.
Chaves M, Albert R, Sontag E (2005) Robustness and fragility of Boolean
models for genetic regulatory networks. J Theor Biol 235: 431–449.
54.
Roelfsema MR, Levchenko V, Hedrich R (2004) ABA depolarizes guard
cells in intact plants, through a transient activation of r- and S-type anion
channels. Plant J 37: 578–588.
55.
Klu¨ sener B, Young JJ, Murata Y, Allen GJ, Mori IC, et al. (2002)
Convergence of calcium signaling pathways of pathogenic elicitors and
abscisic acid in Arabidopsis guard cells. Plant Physiol 130: 2152–2163.
56.
Allen GJ, Chu SP, Harrington CL, Schumacher K, Hoffmann T, et al.
(2001) A defined range of guard cell calcium oscillation parameters
encodes stomatal movements. Nature 411: 1053–1057.
57.
McAinsh MR, Webb A, Taylor JE, Hetherington AM (1995) Stimulus-
induced oscillations in guard cell cytosolic free calcium. Plant Cell 7:
1207–1219.
58.
Jacob T, Ritchie S, Assmann SM, Gilroy S (1999) Abscisic acid signal
transduction in guard cells is mediated by phospholipase D activity. Proc
Natl Acad Sci U S A 96: 12192–12197.
59.
Gehring CA, Irving HR, Parish RW (1990) Effects of auxin and abscisic
acid on cytosolic calcium and pH in plant cells. Proc Natl Acad Sci U S A
87: 9645–9649.
60.
Tena G, Renaudin JP (1998) Cytosolic acidification but not auxin at
physiological concentration is an activator of map kinases in tobacco cells.
Plant J 16: 173–182.
61.
Assmann SM (2004) Plant G proteins, phytohormones, and plasticity:
Three questions and a speculation. Sci STKE 2004: re20.
62.
Hetherington AM, Woodward FI (2003) The role of stomata in sensing and
driving environmental change. Nature 424: 901–908.
63.
Farquhar GD, Cowan IR (1974) Oscillations in stomatal conductance: The
influence of environmental gain. Plant Physiol 54: 769–772.
64.
Cowan IR, Farquhar GD (1977) Stomatal function in relation to leaf
metabolism and environment. Symp Soc Exp Biol 31: 471–505.
65.
Espinosa-Soto C, Padilla-Longoria P, Alvarez-Buylla ER (2004) A gene
regulatory network model for cell-fate determination during Arabidopsis
thaliana flower development that is robust and recovers experimental gene
expression profiles. Plant Cell 16: 2923–2939.
66.
Thum KE, Shasha DE, Lejay LV, Coruzzi GM (2003) Light- and carbon-
signaling pathways. Modeling circuits of interactions. Plant Physiol 132:
440–452.
67.
Genoud T, Metraux JP (1999) Crosstalk in plant cell signaling: Structure
and function of the genetic network. Trends Plant Sci 4: 503–507.
68.
Genoud T, Trevino Santa Cruz MB, Metraux JP (2001) Numeric simulation
of plant signaling networks. Plant Physiol 126: 1430–1437.
69.
Alon U, Surette MG, Barkai N, Leibler S (1999) Robustness in bacterial
chemotaxis. Nature 397: 168–171.
70.
Sokolovski S, Blatt MR (2004) Nitric oxide block of outward-rectifying Kþ
channels indicates direct control by protein nitrosylation in guard cells.
Plant Physiol 136: 4275–4284.
71.
Linder B, Raschke K (1992) A slow anion channel in guard cells, activating
at large hyperpolarization, may be principal for stomatal closing. FEBS
Lett 313: 27–30.
72.
Hosy E, Vavasseur A, Mouline K, Dreyer I, Gaymard F, et al. (2003) The
Arabidopsis outward Kþ channel GORK is involved in regulation of
stomatal movements and plant transpiration. Proc Natl Acad Sci U S A
100: 5549–5554.
73.
Mills LN, Hunt L, Leckie CP, Aitken FL, Wentworth M, et al. (2004) The
effects of manipulating phospholipase C on guard cell ABA-signalling. J
Exp Bot 55: 199–204.
74.
Assmann SM, Grantz DA (1990) Stomatal response to humidity in
sugarcane and soybean: Effect of vapour pressure difference on the
kinetics of the blue light response. Plant Cell Environ 13: 163–169.
75.
Pearcy R, W. (1990) Sunflecks and photosynthesis in plant canopies. Annu
Rev Plant Physiol Plant Mol Biol 41: 421–453.
76.
Jeong H, Mason SP, Baraba´si AL, Oltvai ZN (2001) Lethality and centrality
in protein networks. Nature 411: 41–42.
77.
Said MR, Begley TJ, Oppenheim AV, Lauffenburger DA, Samson LD
(2004) Global network analysis of phenotypic effects: Protein networks
and toxicity modulation in Saccharomyces cerevisiae. Proc Natl Acad Sci U S
A 101: 18006–18011.
78.
Kinney RU, Crucitti P, Albert R, Latora V (2005) Modeling cascading
failures in the North American power grid. Eur Phys J B 46: 101–107.
79.
Sanders D, Pelloux J, Brownlee C, Harper JF (2002) Calcium at the
crossroads of signaling. Plant Cell 14: S401–S417.
80.
Hirschi KD, Zhen RG, Cunningham KW, Rea PA, Fink GR (1996) CAX1, an
Hþ/Ca2þ antiporter from Arabidopsis. Proc Natl Acad Sci U S A 93: 8782–
8786.
81.
Staxen II, Pical C, Montgomery LT, Gray JE, Hetherington AM, et al.
(1999) Abscisic acid induces oscillations in guard-cell cytosolic free
calcium that involve phosphoinositide-specific phospholipase C. Proc
Natl Acad Sci U S A 96: 1779–1784.
82.
Allen GJ, Kwak JM, Chu SP, Llopis J, Tsien RY, et al. (1999) Cameleon
calcium indicator reports cytoplasmic calcium dynamics in Arabidopsis
guard cells. Plant J 19: 735–747.
83.
MacRobbie EA (1998) Signal transduction and ion channels in guard cells.
Philos Trans R Soc Lond B Biol Sci 353: 1475–1488.
84.
Romano LA, Jacob T, Gilroy S, Assmann SM (2000) Increases in cytosolic
Ca2þ are not required for abscisic acid-inhibition of inward Kþ currents in
guard cells of Vicia faba L. Planta 211: 209–217.
85.
Levchenko V, Konrad KR, Dietrich P, Roelfsema MR, Hedrich R (2005)
Cytosolic abscisic acid activates guard cell anion channels without
preceding Ca2þ signals. Proc Natl Acad Sci U S A 102: 4203–4208.
86.
Gilroy S, Fricker MD, Read ND, Trewavas AJ (1991) Role of calcium in
signal transduction of Commelina guard cells. Plant Cell 3: 333–344.
87.
Allan AC, Fricker MD, Ward JL, Beale MH, Trewavas AJ (1994) Two
transduction pathways mediate rapid effects of abscisic acid in Commelina
guard cells. Plant Cell 6: 1319–1328.
88.
Klu¨ sener B, Young JJ, Murata Y, Allen GJ, Mori IC, et al. (2002)
Convergence of calcium signaling pathways of pathogenic elicitors and
abscisic acid in Arabidopsis guard cells. Plant Physiol 130: 2152–2163.
89.
Webb AA, Larman MG, Montgomery LT, Taylor JE, Hetherington AM
(2001) The role of calcium in ABA-induced gene expression and stomatal
movements. Plant J 26: 351–362.
90.
Raschke K, Shabahang M, Wolf R (2003) The slow and the quick anion
conductance in whole guard cells: Their voltage-dependent alternation,
and the modulation of their activities by abscisic acid and CO2. Planta
217: 639–650.
91.
Kwak JM, Moon JH, Murata Y, Kuchitsu K, Leonhardt N, et al. (2002)
Disruption of a guard cell-expressed protein phosphatase 2A regulatory
subunit, RCN1, confers abscisic acid insensitivity in Arabidopsis. Plant Cell
14: 2849–2861.
92.
Mangan S, Alon U (2003) Structure and function of the feed-forward loop
network motif. Proc Natl Acad Sci U S A 100: 11980–11985.
93.
Brandman O, Ferrell JE, Jr., Li R, Meyer T (2005) Interlinked fast and slow
positive feedback loops drive reliable cell decisions. Science 310: 496–498.
94.
Razem FA, El-Kereamy A, Abrams SR, Hill RD (2006) The RNA-binding
protein FCA is an abscisic acid receptor. Nature 439: 290–294.
95.
Glass L, Kauffman SA (1973) The logical analysis of continuous, non-
linear biochemical control networks. J Theor Biol 39: 103–129.
96.
Chaves M, Sontag E, Albert R. (2006) Methods of robustness analysis for
Boolean models of gene control networks. IEE Proc Systems Biology 153:
154–167.
97.
Peak D, West JD, Messinger SM, Mott KA (2004) Evidence for complex,
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1747
Model of Guard Cell ABA Signaling
collective dynamics and emergent, distributed computation in plants.
Proc Natl Acad Sci U S A 101: 918–922.
98.
Arabidopsis Genome Initiative T (2000) Analysis of the genome sequence
of the flowering plant Arabidopsis thaliana. Nature 408: 796–815.
99.
Spiro PA, Parkinson JS, Othmer HG (1997) A model of excitation and
adaptation in bacterial chemotaxis. Proc Natl Acad Sci U S A 94: 7263–
7268.
100. Hoffmann A, Levchenko A, Scott ML, Baltimore D (2002) The IjB-NFjB
signaling module: Temporal control and selective gene activation. Science
298: 1241–1245.
101. Rao CV, Wolf DM, Arkin AP (2002) Control, exploitation and tolerance of
intracellular noise. Nature 420: 231–237.
102. Shen-Orr SS, Milo R, Mangan S, Alon U (2002) Network motifs in the
transcriptional regulation network of Escherichia coli. Nat Genet 31: 64–68.
103. Prill RJ, Iglesias PA, Levchenko A (2005) Dynamic properties of network
motifs contribute to biological network organization. PLoS Biol 3: e343.
DOI: 10.1371/journal.pbio.0030343
104. Bornholdt S (2005) Systems biology. Less is more in modeling large
genetic networks. Science 310: 449–451.
105. Klemm K, Bornholdt S (2005) Topology of biological networks and
reliability of information processing. Proc Natl Acad Sci U S A 102:
18414–18419.
106. Ingolia NT (2004) Topology and robustness in the Drosophila segment
polarity network. PLoS Biol 2: e123. DOI: 10.1371/journal.pbio.0020123
107. Gutterson N, Zhang JZ (2004) Genomics applications to biotech traits: A
revolution in progress? Curr Opin Plant Biol 7: 226–230.
108. Wang W, Vinocur B, Altman A (2003) Plant responses to drought, salinity
and extreme temperatures: Towards genetic engineering for stress
tolerance. Planta 218: 1–14.
109. Zhang JZ, Creelman RA, Zhu JK (2004) From laboratory to field. Using
information from Arabidopsis to engineer salt, cold, and drought tolerance
in crops. Plant Physiol 135: 615–621.
110. Delmer DP (2005) Agriculture in the developing world: Connecting
innovations in plant research to downstream applications. Proc Natl Acad
Sci U S A 102: 15739–15746.
111. Moffat AS (2002) Plant genetics. Finding new ways to protect drought-
stricken plants. Science 296: 1226–1229.
112. Breshears DD, Cobb NS, Rich PM, Price KP, Allen CD, et al. (2005)
Regional vegetation die-off in response to global-change-type drought.
Proc Natl Acad Sci U S A 102: 15144–15148.
113. Schroter D, Cramer W, Leemans R, Prentice IC, Araujo MB, et al. (2005)
Ecosystem service supply and vulnerability to global change in Europe.
Science 310: 1333–1337.
114. Rao BM, Lauffenburger DA, Wittrup KD (2005) Integrating cell-level
kinetic modeling into the design of engineered protein therapeutics. Nat
Biotechnol 23: 191–194.
115. Liu ET, Kuznetsov VA, Miller LD (2006) In the pursuit of complexity:
Systems medicine in cancer biology. Cancer Cell 9: 245–247.
116. Irish JM, Hovland R, Krutzik PO, Perez OD, Bruserud O, et al. (2004)
Single cell profiling of potentiated phospho-protein networks in cancer
cells. Cell 118: 217–228.
117. Irish JM, Kotecha N, Nolan GP (2006) Mapping normal and cancer cell
signalling networks: Towards single-cell proteomics. Nat Rev Cancer 6:
146–155.
118. Krull M, Pistor S, Voss N, Kel A, Reuter I, et al. (2006) Transpath: An
information resource for storing and visualizing signaling pathways and
their pathological aberrations. Nucleic Acids Res 34: D546–D551.
PLoS Biology | www.plosbiology.org
October 2006 | Volume 4 | Issue 10 | e312
1748
Model of Guard Cell ABA Signaling
|
16968132
|
ROS = ( Atrboh )
PEPC = NOT ( ( ABA ) )
PLD = ( GPA1 )
HTPase = NOT ( ( pH ) OR ( Ca2_c ) OR ( ROS ) )
Ca2_c = ( ( CIS ) AND NOT ( Ca2_ATPase ) ) OR ( ( CaIM ) AND NOT ( Ca2_ATPase ) )
ROP10 = ( ERA1 )
RAC1 = NOT ( ( ABA ) OR ( ABI1 ) )
OST1 = ( ABA )
ROP2 = ( PA )
InsP6 = ( InsPK )
SphK = ( ABA )
Depolar = ( ( KOUT AND ( ( ( NOT AnionEM AND NOT Ca2_c AND NOT HTPase AND NOT KEV ) ) ) ) OR ( Ca2_c ) OR ( KEV ) OR ( HTPase AND ( ( ( NOT AnionEM AND NOT Ca2_c AND NOT KOUT AND NOT KEV ) ) ) ) OR ( AnionEM ) ) OR NOT ( AnionEM OR Ca2_c OR HTPase OR KOUT OR KEV )
RCN1 = ( ABA )
Ca2_ATPase = ( Ca2_c )
NOS = ( Ca2_c )
GPA1 = ( ( AGB1 ) AND NOT ( GCR1 ) ) OR ( S1P AND ( ( ( AGB1 ) ) ) )
Atrboh = ( ( OST1 AND ( ( ( pH AND ROP2 ) ) ) ) AND NOT ( ABI1 ) )
Malate = ( ( ( PEPC ) AND NOT ( AnionEM ) ) AND NOT ( ABA ) )
AnionEM = ( pH AND ( ( ( Ca2_c ) ) OR ( ( NOT ABI1 ) ) ) ) OR ( Ca2_c AND ( ( ( pH ) ) OR ( ( NOT ABI1 ) ) ) )
KAP = ( ( Depolar ) AND NOT ( Ca2_c AND ( ( ( pH ) ) ) ) )
pH = ( ABA )
CIS = ( InsP3 AND ( ( ( InsP6 ) ) ) ) OR ( cGMP AND ( ( ( cADPR ) ) ) )
InsP3 = ( PLC )
PA = ( PLD )
ABI1 = ( ( ( pH ) AND NOT ( PA ) ) AND NOT ( ROS ) )
CaIM = ( ( ( ABH1 AND ( ( ( NOT ERA1 ) ) ) ) AND NOT ( Depolar ) ) OR ( ( ERA1 AND ( ( ( NOT ABH1 ) ) ) ) AND NOT ( Depolar ) ) OR ( ( ROS ) AND NOT ( Depolar ) ) ) OR NOT ( ROS OR ERA1 OR ABH1 OR Depolar )
S1P = ( SphK )
NIA12 = ( RCN1 )
cGMP = ( GC )
PLC = ( ABA AND ( ( ( Ca2_c ) ) ) )
cADPR = ( ADPRc )
ADPRc = ( NO )
Actin = ( ( Ca2_c ) ) OR NOT ( RAC1 OR Ca2_c )
AGB1 = ( GPA1 )
Closure = ( ( KOUT AND ( ( ( AnionEM ) ) AND ( ( Actin ) ) ) ) AND NOT ( Malate ) ) OR ( ( KAP AND ( ( ( AnionEM ) ) AND ( ( Actin ) ) ) ) AND NOT ( Malate ) )
InsPK = ( ABA )
KEV = ( Ca2_c )
KOUT = ( pH AND ( ( ( Depolar ) ) ) ) OR ( ( Depolar ) AND NOT ( ROS AND ( ( ( NO ) ) ) ) )
GC = ( NO )
NO = ( NOS AND ( ( ( NIA12 ) ) ) )
|
A Logical Model Provides Insights into T Cell
Receptor Signaling
Julio Saez-Rodriguez1, Luca Simeoni2, Jonathan A. Lindquist2, Rebecca Hemenway1, Ursula Bommhardt2,
Boerge Arndt2, Utz-Uwe Haus3, Robert Weismantel3, Ernst D. Gilles1, Steffen Klamt1*, Burkhart Schraven2*
1 Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany, 2 Institute of Immunology, Otto-von-Guericke University, Magdeburg, Germany,
3 Institute for Mathematical Optimization, Otto-von-Guericke University, Magdeburg, Germany
Cellular decisions are determined by complex molecular interaction networks. Large-scale signaling networks are
currently being reconstructed, but the kinetic parameters and quantitative data that would allow for dynamic
modeling are still scarce. Therefore, computational studies based upon the structure of these networks are of great
interest. Here, a methodology relying on a logical formalism is applied to the functional analysis of the complex
signaling network governing the activation of T cells via the T cell receptor, the CD4/CD8 co-receptors, and the
accessory signaling receptor CD28. Our large-scale Boolean model, which comprises 94 nodes and 123 interactions and
is based upon well-established qualitative knowledge from primary T cells, reveals important structural features (e.g.,
feedback loops and network-wide dependencies) and recapitulates the global behavior of this network for an array of
published data on T cell activation in wild-type and knock-out conditions. More importantly, the model predicted
unexpected signaling events after antibody-mediated perturbation of CD28 and after genetic knockout of the kinase
Fyn that were subsequently experimentally validated. Finally, we show that the logical model reveals key elements
and potential failure modes in network functioning and provides candidates for missing links. In summary, our large-
scale logical model for T cell activation proved to be a promising in silico tool, and it inspires immunologists to ask new
questions. We think that it holds valuable potential in foreseeing the effects of drugs and network modifications.
Citation: Saez-Rodriguez J, Simeoni L, Lindquist JA, Hemenway R, Bommhardt U, et al. (2007) A logical model provides insights into T cell receptor signaling. PLoS Comput
Biol 3(8): e163. doi:10.1371/journal.pcbi.0030163
Introduction
Understanding how cellular networks function in a holistic
perspective is the main purpose of systems biology [1].
Dynamic models provide an optimal basis for a detailed study
of cellular networks and have been applied successfully to
cellular networks of moderate size [2–5]. However, for their
construction and analysis they require an enormous amount
of mechanistic details and quantitative data which, until now,
has been often lacking in large-scale networks. Therefore,
there has been considerable effort to develop methods based
exclusively on the often well-known network topology [6,7].
One may distinguish between studies on the statistical
properties of graphs [8–10] and approaches aiming at
predicting functional or dysfunctional states and modes.
For the latter, a large corpus of methods has been developed
for metabolic networks mainly relying on the constraints-
based approach [11,12]. However, for signaling networks,
methods facilitating a similar functional analysis—including
predictions on the outcome of interventions— have been
applied to a much lesser extent [6].
Here we demonstrate that capturing the structure of
signaling networks by a recently introduced logical approach
[13] allows the analysis of important functional aspects, often
leading to predictions that can be verified in knock-out/
perturbation experiments. Logical networks have until now
been used for studying artificial (random) networks [14] or
relatively small gene regulatory networks [15–18]. In contrast,
herein we study a large-scale signaling network, structured in
input (e.g., receptors), intermediate, and output (e.g., tran-
scription factors) layers. Compared with gene regulatory
networks, the behavior of signaling networks is mainly
governed by their input layer, shifting the interest to input–
output relationships. Addressing these issues requires parti-
ally different techniques, as compared with gene regulatory
networks. We use a special and intuitive representation of
logical networks (called logical interaction hypergraph (LIH); see
Methods), which is well-suited for this kind of input–output
analysis. By applying logical steady state analysis, one may
predict how a combination of signals arriving at the input
layer leads to a certain response in the intermediate and the
output layers. Additionally, this approach facilitates predic-
tions of the effect of interventions and, moreover, allows one
to search for interventions that repress or provoke a certain
logical response [13]. Furthermore, each logical network has a
unique underlying interaction graph from which other
important network properties such as feedback loops,
signaling paths, and network-wide interdependencies can be
evaluated.
Editor: Rob J. De Boer, Utrecht University, The Netherlands
Received February 6, 2007; Accepted July 5, 2007; Published August 24, 2007
A previous version of this article appeared as an Early Online Release on July 5,
2007 (doi:10.1371/journal.pcbi.0030163.eor).
Copyright: 2007 Saez-Rodriguez et al. This is an open-access article distributed
under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the
original author and source are credited.
Abbreviations: LIH, logical interaction hypergraph; MHC, Major Histocompatibility
Complex; MIS, Minimal intervention set; TCR, T cell receptor
* To whom correspondence should be addressed. E-mail: inquiries regarding the
mathematical methodology should be addressed to Steffen Klamt, klamt@
mpi-magdeburg.mpg.de, and regarding the biological and experimental data to
Burkhart Schraven, Burkhart.Schraven@med.ovgu.de
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1580
Importantly, we consider here a logical model to be
constructed by collecting and integrating well-known local
interactions (e.g., a kinase phosphorylates an adaptor
molecule). The logical model is then employed to derive
global information (e.g., stimulation of a receptor leads to the
activation of a certain transcription factor via several logical
connections). Thus, the available data on the global network
behavior is not used to construct the model; instead, it is used
to verify the model. The model may then be employed to
predict global responses that have not yet been studied
experimentally.
Here, we apply the logical framework to a carefully
constructed model of T cell receptor (TCR) signaling. T-
lymphocytes play a key role within the immune system:
cytotoxic, CD8þ, T cells destroy cells infected by viruses or
malignant cells, and CD4þ T helper cells coordinate the
functions of other cells of the immune system [19]. The
importance of T cells for immune homeostasis is due to their
ability to specifically recognize foreign, potentially danger-
ous, agents and, subsequently, to initiate a specific immune
response. T cell reactivity must be exquisitely regulated as
either a decrease (which weakens the defense against
pathogens with the consequence of immunodeficiency) or
an increase (which can lead to autoimmune disorders and
leukemia) can have severe consequences for the organism.
T cells detect foreign antigens by means of the TCR, which
recognizes peptides only when presented upon MHC (Major
Histocompatibility Complex) molecules. The peptides that
are recognized by the TCR are typically derived from foreign
(e.g., bacterial, viral) proteins and are generated by proteo-
lytic cleavage within so-called antigen presenting cells (APCs).
Binding of the TCR to peptide/MHC complexes and the
additional binding of a different region of the MHC
molecules by the co-receptors (CD4 in the case of T helper
cells and CD8 in the case of cytotoxic T cells), together with
costimulatory molecules such as CD28, initiates a plethora of
signaling cascades within the T cell. These cascades give rise
to a complex signaling network, which controls the activation
of several transcription factors. These transcription factors,
in turn, control the cell’s fate, particularly whether the T cell
becomes activated and proliferates or not [20]. Therefore, we
chose to focus on a limited number of receptors that are
known to be central to the decision making process. The high
number of kinases, phosphatases, adaptor molecules, and
their interactions give rise to a complex interaction network
which cannot be interpreted via pure intuition and requires
the aid of mathematical tools. Since no sufficient basis of
kinetic data is available for setting up a dynamic model of this
network, we opted to use logical modeling as a qualitative and
discrete modeling framework. Note that there are kinetic
models dealing with a smaller part of the network (e.g.,
[5,21,22]), as well as models of the gene regulatory network
governing T cell activation [23].
We recently introduced our approach for the logical
modeling of signaling networks [13], and, to exemplify it, we
presented a small logical model for T cell activation (40
nodes). However, this model only served to demonstrate
applicability and was too incomplete to address realistic
complex input–output patterns. In contrast, the model
presented herein has been significantly expanded to 94 nodes
and refined by a careful reconstruction process (see below). It
is thus realistic enough to be verified with diverse exper-
imental data and to test its predictive power.
In this report, the large-scale logical model describing T
cell activation and the analysis performed therewith will be
presented. First we will show that a number of important
structural features can be identified with this model. Then we
will show that the model not only reproduces published data
on wet lab experiments, but it also predicts non-intuitive and
previously unknown responses.
Results
Setup of a Curated, Comprehensive Logical Model of T
Cell Receptor Signaling
We have constructed a logical model describing T cell
signaling (see Methods and Figure 1), which comprises the
main events and elements connecting the TCR, its corecep-
tors CD4/CD8, and the costimulatory molecule CD28, to the
activation of key transcription factors in T cells such as AP-1,
NFAT, and NFjB, all of which determine T cell activation and
T cell function. In general, the model includes the following
signaling steps emerging from the above receptors: the
activation of the Src kinases Lck and Fyn, followed by the
activation of the Syk-related protein tyrosine kinase ZAP70,
and the subsequent assembly of the LAT signalosome, which
in turn triggers activation of PLCc1, calcium cascades,
activation of RasGRP, and Grb2/SOS, leading to the activa-
tion of MAPKs [20]. Additionally, it includes the activation of
the PI3K/PKB pathway that regulates many aspects of cellular
activation and differentiation, particularly survival. For the
activation of elements that play an important role, but whose
regulation is not well-known yet (e.g., Card11, Gadd45), an
external input was added. These elements can be considered
as points of future extension of the model.
As mentioned above, our model, which is documented in a
detailed manner in Tables S1 and S2, is based upon local
interactions (e.g., kinase ZAP70 phosphorylates the adaptor
molecule LAT) that are well-established for primary T cells in
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1581
Author Summary
T-lymphocytes are central regulators of the adaptive immune
response, and their inappropriate activation can cause autoimmune
diseases or cancer. The understanding of the signaling mechanisms
underlying T cell activation is a prerequisite to develop new
strategies for pharmacological intervention and disease treatments.
However, much of the existing literature on T cell signaling is related
to T cell development or to activation processes in transformed T
cell lines (e.g., Jurkat), whereas information on non-transformed
primary T cells is limited. Here, immunologists and theoreticians
have compiled data from the existing literature that stem from
analysis of primary T cells. They used this information to establish a
qualitative Boolean network that describes T cell activation
mechanisms after engagement of the TCR, the CD4/CD8 co-
receptors, and CD28. The network comprises 94 nodes and can be
extended to facilitate interpretation of new data that emerge from
experimental analysis of T cell activation. Newly developed tools and
methods allow in silico analysis, and manipulation of the network
and can uncover hidden/unforeseen signaling pathways. Indeed, by
assessing signaling events controlled by CD28 and the protein
tyrosine kinase Fyn, we show that computational analysis of even a
qualitative network can provide new and non-obvious signaling
pathways which can be validated experimentally.
A Logical Model of T Cell Receptor Signaling
the literature. We did not use the known global information
(e.g., stimulation of a receptor leads to the activation of a
certain transcription factor) for the model construction.
Instead, in simulations, the local interactions give rise to a
global behavior which can be compared with available
experimental observations (and was thus used to verify the
model).
Each component in the logical model can be either ON
(‘‘1’’) or OFF (‘‘0’’). We consider a compound to be ON only if
it is fully activated and able to trigger downstream events
properly; otherwise, it is OFF. Furthermore, we consider two
timescales [13]: early (s ¼ 1) and late (s ¼ 2), involving
processes occurring during or after the first minutes of
activation, respectively (the time-scale for each interaction is
given in Table S2). Some key regulatory processes such as the
degradation of signaling proteins mediated by the E3
ubiquitin ligase c-Cbl [24–26] occur after a certain time,
and are thus assigned s ¼ 2. Therefore, as will be shown later,
analysis of signal propagation during the early events reveals
which elements become activated, and the consideration of
the late events allows a rough approximation to the dynamic
behavior (sustained versus transient) of the network.
The model comprises 94 different compounds and 123
interactions that give rise to a complex map of interactions
(Figure 1). It is, to the best of our knowledge, the largest
Boolean model of a cellular network to date.
Interaction-Graph-Based Analyses
The first step in our analysis was to examine the interaction
graph underlying the logical model. The former can be easily
derived from the latter when a special representation of
Boolean networks is used (see Methods). The interaction
graph is less constrained than the Boolean network since it
only captures direct (positive or negative) effects of one
Figure 1. Logical Model of T Cell Activation (Screenshot of CellNetAnalyzer)
Each arrow pointing at a species box is a so-called hyperarc representing one possibility to activate that species (see Methods). All the hyperarcs pointing
at a particular species box are OR connected. Yellow species boxes denote output elements, while green ones represent (co)receptors. In the shown
‘‘early-event’’ scenario, the feedback loops were switched off, and only the input for the costimulatory molecule CD28 is active (scenario in column 2 of
Table 1). The resulting logical steady state was then computed. Small text boxes display the signal flows along the hyperarcs (blue boxes: fixed values
prior to computation; green boxes: hyperarcs activating a species (signal flow is 1); red boxes: hyperarcs which are not active (signal flow is 0)).
doi:10.1371/journal.pcbi.0030163.g001
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1582
A Logical Model of T Cell Receptor Signaling
molecule upon another. Thus, unlike the logical model, the
interaction graph cannot describe how different causal
effects converging at a certain species are combined. For
example, in an interaction graph we may say that A and B
have a positive influence on another node C; the logical
network is more precise because it expresses that A AND B
(or A OR B) are required to activate C. Accordingly,
compared with the logical model, an interaction graph
requires less a priori knowledge about the network under
study which comes at the price that functional predictions
are limited. Nevertheless, as demonstrated in this section, a
number of important functional features can be revealed
from the graph model.
First we studied global properties of the graph. As
expected, the graph is connected (i.e., neglecting the arc
directions, there is always a path from one node to all others).
However, the directed graph contains as a core one strongly
connected component with 33 nodes (i.e., for each pair (a,b)
of nodes taken from this component there is a path from a to
b and from b to a). This structural organization is related with
the bow-tie structure found in other cellular networks (e.g.,
[7,27]) and implies that the rest of the network (not contained
in the strongly connected component) mainly consists in
relatively simple input and output layers (including branch-
ing cascades) feeding to and from this component.
We continued the interaction-graph-based analysis by
computing the feedback loops. Feedback loops are of major
importance for the dynamic behavior and functioning of
biological networks. Negative feedback loops control homeo-
static response and can give rise to oscillations, while positive
feedbacks govern multistable behavior (connected to irrever-
sible decision-making and differentiation processes) [15,28–
30]. The interaction graph underlying the logical T cell model
has 172 feedback loops, 89 thereof being negative. Remark-
ably, all feedback loops are only active in the second timescale
because each loop contains at least one process of the second
timescale. The elements of the MAPK cascade are involved in
92% of the feedback loops. This is due to the fact that there is
a connection from ERK to the phosphatase SHP1 from the
bottom to the top of the network [5]. Due to this connection,
the resulting feedback can return to ERK via many different
paths, thereby leading to a high number of loops. Indeed, if
the ERK ! SHP1 connection is not considered, the number
of loops is reduced dramatically from 172 to 13 (with only 11
being negative), all located in the upper part of the network.
c-Cbl is involved in ;85% of them, thus underscoring the
importance of c-Cbl in the regulation of signaling processes
[25,26].
There are 4,538 paths, each connecting one of the three
compounds from the input layer (TCR, CD4/CD8, CD28) with
one compound in the output layer (transcription factors and
other elements controlling T cell activation). The high
number of negative paths (2,058) can be traced back to the
presence of two negative connections (via DGK and Gab2). In
fact, considering the early signaling events within the
network, where DGK and Gab2 are not active yet, the
number of paths is reduced to 1,530, with only six of them
being negative. These paths are from the TCR and CD28 to
negative regulators of the cell cycle (p21, p27, and FKHR),
having thus a positive effect on T cell proliferation. These
and other global effects can be graphically inspected via the
dependency matrix [13,31], depicted in Figure 2. Importantly,
when considering the timescale s ¼ 1, there is no ambivalent
effect (i.e., via positive and negative paths) between any
ordered pair (A,B) of species, i.e., A is either a pure activator
of B (only positive paths from A to B), or a pure inhibitor of B
(only negative paths from A to B), or has no direct or indirect
influence on B at all. For example, during early activation, the
TCR can only have a positive effect upon AP1 (the array
element (TCRb, AP1) in Figure 2 is green). Note that this
changes for timescale s ¼ 2 where, in several cases, a
compound influences another species in an ambivalent
manner.
Analysis of the Logical Model
An important aspect that can be studied with a logical
model is signal processing and signal propagation and the
corresponding response (activation/inactivation) of the nodes
upon external stimuli and perturbations (see Methods). One
starts the analysis of a scenario by defining a pattern of input
stimuli, possibly in combination with a set of nodes that are
knocked-out or knocked-in. Then, by an iterative evaluation
of the Boolean rules in each node, the signal is propagated
through the network, switching each node ON or OFF,
respectively (see [13] and Methods). For example, since CD28
(an input) is (permanently) ON in the scenario shown in
Figure 1, it will (permanently) activate node X, which will in
turn (permanently) activate Vav1, and so forth. In the same
scenario, since the input CD4 is OFF, Lckp1 and therefore
Abl, ZAP70, and other components cannot become activated
and therefore are in the OFF state. In the ideal case, each
node can be assigned a uniquely determined state that follows
from a given input pattern. In terms of Boolean networks, the
set of determined node values then represents a logical steady
state. In some cases, in particular when negative feedback
loops are active, only a fraction of the elements can be
assigned a unique steady state value, whereas other (or even
all) nodes might oscillate [15]. However, since in the T cell
model all negative feedback loops become active only during
timescale s ¼ 2 (as described above), a complete logical steady
state follows for arbitrary input patterns when considering s
¼ 1.
Using this kind of logical steady state analysis, we first
analyzed the activation pattern of key elements upon differ-
ent stimuli (activation of the TCR and/or CD4 and/or CD28;
Table 1) for timescale s ¼ 1. The model was able to reproduce
data from both the literature and our own experiments,
providing a holistic and integrated interpretation for a large
body of data. The model also predicted a non-obvious
signaling event, namely that the activation of the costimula-
tory molecule CD28 alone leads not only to the activation of
PI3K—which is to be expected from a large body of literature
dealing with CD28 signaling showing that PI3K binds to the
motif YxxM of CD28 [32,33]—but also to the selective
activation of JNK, but not ERK. The model predicts a
pathway from CD28 to JNK which gives a holistic explanation
for this result: the pathway does NOT involve the LAT
signalosome, activation of PLCc1, and Calcium flux, but
clearly depends on the activation of the nucleotide exchange
factor Vav1 which activates MEKK1 via the small G-protein
Rac1 (Figure 1). Clearly, the activating pathway shown in
Figure 1 could be identified by a visual inspection of the map
(note that we have intentionally drawn the network in such a
way that this route can be easily seen). However, in large-scale
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1583
A Logical Model of T Cell Receptor Signaling
networks the identification of long-distance pathways by
simply following all possible routes becomes infeasible and is
particularly complicated if AND connections are involved.
Furthermore, since the CD28-induced JNK activation path-
way was not expected, one would probably not have searched
specifically for this pathway, while the algorithm reveals the
whole response of the network.
The prediction made by the model is particularly surpris-
ing in light of published data which either suggest that CD28
stimulation alone does NOT activate JNK [34,35] or induces
only a weak activation [36]. Driven by this surprising
prediction, we performed the corresponding experiments in
vitro. As shown in Figure 3A, stimulation of mouse primary T
cells with a non-superagonistic CD28 antibody induced an
evident and sustained JNK phosphorylation, thus confirming
almost perfectly the predicted binary response. Note, the
model also predicted that JNK activation does not depend on
the activation of PI3K. Again, this prediction was verified by
applying a pharmacological inhibitor of PI3K (Figure 3D).
The discrepancies with the literature could be due either to
the different cellular systems (primary T cells versus T cell
lines) or to the different stimulation conditions.
The nature of the kinase involved in CD28-mediated
signaling remains unclear. Indeed, application of the Src-
kinase inhibitor PP2 that inactivates both Lck þ Fyn [37],
showed that Src-kinases, which were proposed to mediate
CD28 signaling [38], are dispensable for the CD28-mediated
activation of JNK (Figure 4). To fit the Src-kinase inhibitor
data with the model, it would have been possible to simply
bypass the Src-kinase and to draw a causal connection from
CD28 to Vav. Such a connection would indeed be justified
since it is well established that triggering of CD28 leads to the
activation of Vav ([39]; for more details, see Table S2,
reactions 35 and 48). However, we preferred to include a
to-be-identified kinase X that gets activated by CD28 (Figure
1), in order to keep within the model the information that
there is a component to be identified. Potential candidates
for kinase X would be members of the Tec-family of PTKs.
However, it is difficult to study the signaling properties of
these kinases in primary non-transformed cells since specific
inhibitors for Tec kinases are not yet available and the
corresponding knock-out mice show defects in thymic
development. Therefore, as we focused during model
generation on well-established data from primary T cells
and excluded data obtained from knockout mice showing
alterations of thymic development, we did not include it.
Figure 2. Dependency Matrix of the Logical T Cell Signaling Model (Figure 1) for the Early Events Scenario (s ¼ 1)
The color of a matrix element Mxy has the following meaning [13]: (i) dark green: x is a total activator of y; (ii) dark red: x is a total inhibitor of y; (iii) white:
no (direct or indirect) influence from x on y.
doi:10.1371/journal.pcbi.0030163.g002
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1584
A Logical Model of T Cell Receptor Signaling
The ability of the model to recapitulate the T cell
phenotype of a variety of previously described knock-out
mice was also tested (Table 1). Indeed, the model could
reproduce the phenotype of several knock-outs and again
reported a rather unexpected result: activation of the TCR in
Fyn-deficient cells selectively triggers the PI3K/PKB pathway.
This prediction was subsequently tested using peripheral
primary T cells prepared from spleen of Fyn-deficient mice.
As shown in Figure 3B, the wet-lab experiments corroborated
the model result again.
However, there was an experimental result which the
model could not reproduce: TCR-mediated JNK activation is
blocked by an inhibitor of PI3K (Figure 3C). In fact, this result
is not in accordance with the network because PI3K has no
influence upon JNK (see dependency matrix, Figure 2).
To identify potential connections that would explain the
experimental data, we applied the concept of Minimal
Intervention Sets (MISs; see Methods). A MIS is a irreducible
collection of actions (e.g., activation or inactivation of certain
compounds), that, if applied, guarantees that a certain goal (a
desired behavior) is fulfilled [13]. Here, we computed the MISs
by which JNK becomes activated under the experimentally
obtained constraint (see Figure 3C) that PI3K is OFF
(describing the effect of the PI3K inhibitor), ZAP70 is ON,
and that the TCR has been activated. These MISs (Table 2)
thus provide a list of minimal combinations of elements that
should be directly or indirectly affected by PI3K and thus
allow us to explain the observed response of JNK upon
inhibiting PI3K. Some of them are obvious, e.g., the first MIS
in Table 2 suggests that JNK activation could be directly
interacting with PI3K or elements that are located down-
stream of PI3K (e.g., PIP3). There is currently no convincing
experimental evidence for an effect of PI3K on JNK, though.
Other MISs in Table 2 suggest that a PI3K-mediated activation
of Vav (both 1 and 3 isoforms) is involved, which would be an
attractive possibility to explain the experimental data.
Indeed, Vav possesses a PH domain which can bind to PIP3,
and this mechanism could be important for Vav activation
[40], thus making it a reasonable extension of the model.
Another molecule that could be involved in PI3K-mediated
Table 1. Summary of Predicted Activation Pattern upon Different Stimuli and Knock-Out Conditions
Input/
Output
WT
WT
WT
PI3K
PI3K
PI3K
SLP76
Fyn
Fyn
Fyn
Rlk and
Itk
Lck and
Fyn
Lck and
Fyn
Lck and
Fyn
Input
TCR
1
0
1
1
0
1
1
1
1
1
1
1
0
1
CD4
0
0
0
0
0
0
0
0
1
0
0
0
0
0
CD28
0
1
1
0
1
1
0
0
0
1
0
0
1
1
Output
ZAP
1
0
1
1
0
1
1
0
1
0
1
0
0
0
LAT
1
0
1
1
0
1
1
0
1
0
1
0
0
0
PLCga
1
0
1
0
0
0
0
0
1
0
0
0
0
0
ERK
1
0
1
0
0
0
0
0
1
0
0
0
0
0
JNK
1
1
1
1
1
1
1
0
1
1
1
0
1
1
PKB
1
1
1
0
0
0
1
1
1
1
1
0
1
1
AP1
1
0
1
0
0
0
0
0
1
0
0
0
0
0
NFKB
1
0
1
0
0
0
0
0
1
0
0
0
0
0
NFAT
1
0
1
0
0
0
0
0
1
0
0
0
0
0
Reference
Figure
3A, 3C
Figure
3A, 3D
Figures
3A, 4
Figure 3C
Figure 3D
Figure 4
[49]
Figure
3B, [50]
Figure
3B, [50]
Figure
3B, [50]
[51]
Figure 4
Figure 4
Figure 4
The headings denote the perturbed (switched-off) element. In the case of PI3K and Lck and Fyn, the perturbation was done via a chemical inhibitor, and for the rest it was through a
genetic knock-out. The ‘‘Input’’ rows show the stimuli, and ‘‘Output’’ the predictions of the model for key elements of the network. Here, blue numbers denote results corroborated by
published data, while green ones were confirmed by our own data. The red number shows a discrepancy between model and experiment (see discussion in the main text). Finally, the row
labeled Reference indicates the Figure where the experimental results are shown or points to the literature reference.
doi:10.1371/journal.pcbi.0030163.t001
Figure 3. In Vitro Analysis of Model Predictions
(A) Activation of ERK and JNK upon CD28, TCR (CD3), or TCR þ CD28 stimulation in mouse splenic T cells.
(B) Activation of PKB upon TCR, TCRþ CD4, and TCR þ CD28 stimulation in Fyn-deficient and heterozygous splenic mouse T cells.
(C) Inhibition of PI3K with both Ly294002 and Wortmannin blocks the phosphorylation of PKB, ERK, and JNK, but not ZAP-70 in human T cells.
(D) Inhibition of PI3K with both Ly294002 and Wortmannin blocks the phosphorylation of PKB, but not of JNK in human T cells upon CD28 stimulation.
As a control, the total amount of ZAP70 (A) or b-actin (B–D) was determined. One representative experiment (of three) is shown.
doi:10.1371/journal.pcbi.0030163.g003
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1585
A Logical Model of T Cell Receptor Signaling
activation of JNK is the serine/threonine kinase HPK1 (see
Figure 1 and Tables S1 and S2). Interestingly, HPK1 is
phosphorylated by Protein Kinase D1 (PKD1) [41], a kinase
whose activation depends on PKC (which in turn is depend-
ent on DAG, downstream of PI3K) for activation. Since the
regulation and functional roles of both PKD1 and PKC (with
the exception of the h isoform) are not yet well-established in
T cells, we did not include them in the model, but a
connection PI3K ! PIP3 ! Itk ! PLCc ! DAG ! PKC !
PKD1 ! HPK1 would be plausible (in which the path from
PKC to HPK1 via PKD1 would be new). An alternative could
be a Rac-dependent activation of HPK1 [42]; however, this is
again a not-well-established connection and thus was not
considered.
Definitely, the model requires a direct or indirect
connection from PI3K to JNK, and additional experiments
are required to assess which of the candidate links predicted
by the MISs are relevant in peripheral T cells. This particular
example illustrates another useful and important application
of our approach: the model not only reveals that a link is
missing, but also suggests candidates that can be verified
experimentally. Thus, MIS analysis is capable of guiding the
experimentalist and helps to plan the corresponding experi-
ments.
As an additional application of MISs, we computed
combinations of failures (constitutive activation or inactiva-
tion of elements caused for example by mutations) which lead
to sustained T cell activation without external stimuli. These
failure modes would cause uncontrolled proliferation and
thus may be connected to diseases such as leukemia or
autoimmunity. Interestingly, components occurring in the
MISs with few elements (Table 3) are in fact known
oncogenes: ZAP70 [43], PI3K [44], Gab2 [45], and PLCc1
[46] (and SLP76 is directly involved in PLCc1 activation).
Robustness and Sensitivity Analysis of the Logical Model
Strongly related to the idea of MISs is a systematic
evaluation of the network response if the model is confronted
with failures. By considering a failure as something that
happens to the cell by an internal or external event (e.g., a
mutation), we may assess the robustness—one of the most
important properties of living systems [47]—of the network.
In contrast, if we consider the failure as an error that has
been introduced during the modeling process (due to
incomplete knowledge), then we are assessing the sensitivity
of the model with respect to the predictions it makes.
Accordingly, to study robustness and sensitivity issues, we (i)
removed systematically each single interaction from the
network, (ii) recomputed the scenarios given in Table 1, and
(iii) compared the new predictions with the 126 original
predictions (Table 1), ranking the interactions according to
the number of introduced changes produced (Table 4). As an
average value, 4.76 errors were introduced per simulated
failure, which corresponds to 3.78% of the total numbers of
predictions. The most sensitive interactions are mainly
located in the upper part of the network and activate
components such as the T cell receptor (TCRb), ZAP70,
LAT, Fyn, or Abl. It is intuitively clear that the network is very
Figure 4. In Vitro Analysis of Src-Kinase Inhibition
Inhibition of Src-Kinases (Lck and Fyn) with PP2 blocks TCR-induced but affects only moderately CD28-induced PKB and JNK activation in human T cells;
therefore, we concluded that CD28 signaling is not strictly Src-kinase– dependent. The effect was compared with PI3K inhibition via Wortmannin (ccf.
Figure 3C and 3D), which blocks the phosphorylation of PKB but not of JNK. b-actin was included as the loading control. One representative experiment
(of three) is shown.
doi:10.1371/journal.pcbi.0030163.g004
Table 2. Application of the Minimal Intervention Sets To Identify
Candidates To Fill the Gap between PI3K and JNK
MIS
jnk
hpk1
rac1r
hpk1
sh3bp2
mekk1
mkk4
mekk1
mlk3
hpk1
mekk1
rac1p1
hpk1
mekk1
vav1
hpk1
mkk4
rac1p2
hpk1
mlk3
vav3
hpk1
rac1p1
rac1p2
hpk1
rac1p1
vav3
hpk1
rac1p2
vav1
hpk1
vav1
vav3
hpk1
mlk3
rac1p2
The MISs of maximal size 3 to obtain JNK off under the conditions (i) TCR on, (ii) PI3K off,
and (iii) ZAP70 on (as shown in the experiment, see Figure 3D and Table 1) were
computed, setting the rest of conditions to the standard values for the early events. Here,
each MIS represents one set of molecules that should be influenced by PI3K in order to be
consistent with the fact that PI3K inhibition blocks JNK activation. For species
abbreviations, see Tables S1 and S2.
doi:10.1371/journal.pcbi.0030163.t002
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1586
A Logical Model of T Cell Receptor Signaling
sensitive to failures (again, caused either by internal/external
events or modeling errors) in these upper nodes because all
pathways branching downstream are governed by them.
Accordingly, the validation of our model (with the data from
Table 1) is most sensitive to modeling errors in the upper part
of the network. We also note that species that can be
activated by more than one interaction (e.g., PI3K) are
significantly less sensitive to single interaction failures since
alternative pathways exist. Regarding robustness, it is worth
emphasizing that in the worst case about 30% of the original
predictions are affected after removal of an interaction,
indicating that there is no ‘‘all-or-nothing’’ interaction in the
network.
We have also performed the same analysis for the removal
of a species (instead of an interaction) which basically led to
the same results (unpublished data). However, the removal of
a node can be seen as a stronger intervention in the network
than deleting an interaction, as the former simulates the
simultaneous removal of all interactions pointing at that
species. Accordingly, deleting nodes implies some stronger
deviations from the original predictions.
Qualitative Description of the Dynamics
So far we have analyzed which elements within the
signaling network get activated upon signal triggering (i.e.,
for the first timescale s ¼ 1). This is due to the fact that a large
corpus of data for these conditions is available (see Table 1).
However, it is important to note that the model is also able to
roughly predict the dynamics upon different stimuli and
conditions.
The modus operandi goes as follows: first, one computes
the steady state values with no external input (s ¼ 0).
Subsequently, the steady state for s ¼ 1 is computed as
described above. Finally, one computes the state of the ‘‘slow’’
interactions (those only active at s ¼ 2) as a function of the
values at s¼1, and subsequently recomputes the steady states.
This provides the response at late events, s ¼ 2. The results
obtained can be plotted in a time-dependent manner (Figure
5). Here, one can also investigate the effect of different
knock-outs. For example, the absence of PAG has no effect on
key downstream elements of the cascade, due to the
redundant role of other negative regulatory mechanisms
(specifically, the degradation via c-Cbl and Cbl-b, and Gab-2–
mediated inhibition of PLCc1). Only a multiple knock-out of
these regulatory molecules leads to sustained activation of
key elements. Thus, these results point to a certain degree of
redundancy in negative feedbacks for switching off signaling.
This sort of qualitative analysis of the dynamics shows the
ability of the Boolean approach to reproduce the key
dynamic properties (transient versus sustained) of a signaling
process.
Discussion
In this contribution, a logical model describing a large
signaling network was established and analyzed. We set up a
Table 4. Robustness Analysis: Ranked List of the Most Sensitive
Interactions
Interaction
Caused Errors
if Removed
!ccblp1 þ tcrlig ! tcrb
39
!ccblp1 þ tcrp þ abl ! ¼ zap70
34
zap70 ! lat
27
tcrb þ lckr ! fyn
26
tcrbþfyn ! tcrp
26
fyn ! abl
26
pi3k þ !ship1 þ !pten ! pip3
21
lat ! plcgb
15
zap70 þ !gab2 þ gads ! slp76
15
lat ! gads
15
pip3 ! pdk1
13
lckp2 þ !cblb ! pi3k
11
lckr þ tcrb ! lckp2
11
!ikkab ! ikb
11
zap70 þ lat ! sh3bp2
10
plcgb þ !ccblp2 þ slp76 þ zap70 þ vav1 þ itk ! plcga
10
pdk1 ! pkb
10
pip3 þ zap70 þ slp76 ! itk
10
zap70 þ sh3bp2 ! vav1
10
!dgk þ plcga ! dag
9
!shp1 þ cd45 þ cd4 þ !csk þ lckr ! lckp1
8
cd28 ! x
8
tcrb þ lckp1 ! tcrp
8
lckp1 ! abl
8
mek ! erk
6
ras ! raf
6
ca ! cam
6
dag ! rasgrp
6
ip3 ! ca
6
lat ! grb2
6
grb2 ! sos
6
plcga ! ip3
6
raf ! mek
6
sos þ !gap þ rasgrp ! ras
6
x ! vav1
5
mkk4 ! jnk
5
mlk3 ! mkk4
5
rac1p1 ! mlk3
5
rac1r þ vav1 ! rac1p1
5
Each single non-input interaction was removed from the network followed by a
recomputation of the scenarios given in Table 1. The number of deviations from the 126
predictions made in Table 1 is shown. For abbreviations and comments on the
interactions, see Tables S1 and S2.
doi:10.1371/journal.pcbi.0030163.t004
Table 3. Minimal Intervention Sets To Produce the Full
Activation Pattern in T Cells
MIS
!gab2
pi3k
zap70
!gab2
pip3
zap70
pi3k
plcga
zap70
pi3k
slp76
zap70
pip3
slp76
zap70
pip3
plcga
zap70
pdk1
plcga
zap70
The MISs of maximal size 3 that induce sustained full activation (namely: ap1, bcat, bclxl,
cre, cyc1, nfkb, p70s, sre, and nfat are on, whereas fkhr, p21c, and p27k are off) of T cells
without external stimuli. The MISs were computed using CellNetAnalyzer. Note that the
exclamation mark ‘‘!’’ denotes ‘‘deactivation’’; species without this symbol have to be
activated (constitutively). Interestingly, the compounds involved in these MISs are
involved in oncogenesis (ZAP70, PI3K, Gab2, and PLCc1 are oncogenes, and SLP76 is
directly involved in PLCc1 activation, see Figure 1 and main text). Note that since PIP3 is a
second messenger and not ‘‘mutable’’, for the purpose of this analysis the MISs involving
its activation can be considered equivalent to those involving its activator PI3K (i.e., these
MISs are equivalent).
doi:10.1371/journal.pcbi.0030163.t003
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1587
A Logical Model of T Cell Receptor Signaling
comprehensive Boolean model describing T cell signaling and
performed logical steady state analyses unraveling the
processing of signals and the global input–output behavior.
Moreover, by converting the logical model into an interaction
graph, we extracted further important features, such as
feedback loops, signaling paths, and network-wide interde-
pendencies. The latter can be captured in a dependency
matrix (as in Figure 2) which provides thousands of
qualitative predictions that can be falsified in perturbation
experiments. The logical model reproduces the global
behavior of this complex network for both natural and
perturbed conditions (knock-outs, inhibitors, mutations, etc.).
Its validity has been proven by reproducing published data
and by predicting unexpected results that were then verified
experimentally. Table 1 summarizes the results of 14 different
scenarios, in which the logical model predicted 126 states. For
44 of them, experimental data was available (15 from
literature and 29 from our own experiments) confirming
the predictions, except in the case discussed above.
Furthermore, we clearly show that the concept of inter-
vention sets allows one (a) to identify missing links in the
network, (b) to reveal failure modes that can explain the
effects of a physiological dysfunction or disease, and (c) to
search for suitable intervention strategies, while keeping
track of potential side effects, which is valuable for drug
target identification.
Compared with a kinetic model based on differential
equations, a Boolean approach is certainly limited regarding
the analysis of quantitative and dynamical aspects, and it
certainly cannot answer the same questions. However, to
establish such a model requires mainly the topology and only
a relatively small amount of quantitative data; hence, a
combination of information which is currently available in
large-scale networks. Although the model itself is qualitative
(i.e., discrete), it enables us not only to study qualitative
aspects of signaling networks, but it can also be validated by
semi-quantitative measurements such as those in Figures 3
and 4. In summary, with the network involved in T cell
activation as a case study, our approach proved to be a
Figure 5. Considering Different Time Scales, a Rough Description of the Dynamics Can Be Obtained
The activation of key elements upon activation of the TCR, the coreceptor CD4, and the costimulatory molecule CD28 is represented at the resting state,
s ¼ 0 (no inputs); early events s ¼ 1 (input(s), no feedback loops); and later-time events, s ¼ 2 (input(s), feedback loops). The black lines correspond to a
wild type while the green ones to a PAG KO. Note that the absence of PAG has no effect on key downstream elements of the cascade, due to the
redundant role of other negative regulatory mechanisms (degradation via c-Cbl and Cbl-b, Gab-2 mediated inhibition of PLCc1). Multiple knock-out of
these regulatory molecules leads to sustained activation of key elements (red lines).
doi:10.1371/journal.pcbi.0030163.g005
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1588
A Logical Model of T Cell Receptor Signaling
promising in silico tool for the analysis of a large signaling
network, and we think that it holds valuable potential in
foreseeing the effects of drugs and network modifications.
Although sometimes the results of a logical model may
(afterward) appear to be obvious (as in the case of the CD28-
mediated JNK connection), it enables an exhaustive and
rigorous analysis of the information processing taking place
within a signaling network. Such a systematic analysis
becomes infeasible for a human being in large-scale systems.
In addition, the LIH can represent the situation of varying
cofactor functions; for example, that two substances A AND B
are required to activate a third substance C, but activation of
C in the presence of A and a fourth substance D requires B
not to be present.
Certainly, the logical model for T cell activation is far from
complete. We are just at the beginning of the reconstruction
process and other receptors and their pathways need to be
included. However, we feel that already in its current state,
the model may prove useful to inspire immunologists to ask
new questions which may first be answered in silico.
Furthermore, the model may also provide a framework for
those who may endeavor to quantitatively model TCR
signaling.
Methods
Logical network representation and analysis. We began construc-
tion of the signaling network for primary T cells by collecting data
from the literature and from our own experiments providing well-
established connections (Tables S1 and S2). As a first (intermediate)
result, we obtain an interaction graph. Interaction graphs are signed
directed graphs with the molecules (such as receptor, phosphatase, or
transcription factor) as nodes and signed arcs denoting the direct
influence of one species upon another, which can either be activating
(þ) or inhibiting (). For example, a positive arc leads from MEK to
ERK because the first phosphorylates and thereby activates the
second (Figure 1). From the incidence matrix of an interaction graph
we can identify important features such as feedback loops as well as
signaling paths and network-wide interdependencies between pairs
of species (e.g., perturbing A may have no effect on B as there is no
path connecting A to B). Algorithms related to these analyses are
well-known [48] and were recently presented in the context of
signaling networks [13]. However, from interaction graphs we cannot
conclude which combinations of signals reaching a species along the
arcs are required to activate that species. For example, in Figure 1,
Jun AND Fos are required to form active AP1.
For a refined representation of such relationships, we use a logical
(or Boolean) model in which we introduce discrete states for the
species (here the simplest (binary) case: 0 ¼ inactive or not present; 1
¼ active or present) and assign to each species a Boolean function.
Here we use a special representation of Boolean functions known as
disjunctive normal form (DNF, also called ‘‘sum of product’’
representation) which uses exclusively AND, OR, and NOT oper-
ators. A Boolean network with Boolean functions in disjunctive
normal form can be intuitively drawn and stored as a hypergraph
(LIH) [13], which is well-suited for studying the information flows
and input–output relationships in signal transduction networks
(Figure 1). In this hypergraph, each hyperarc connects its start nodes
with an AND operation (indicated by a blue circle in Figure 1) and
each hyperarc represents one possibility for how its end node can be
activated or produced (note that hyperacs may also have only one
start node, i.e., they are then ‘‘graph-like’’ arcs). Red branches
indicate species that enter the hyperarc with their negated value. For
example, PLCc-1 (PLCga in Figure 1) AND NOT DGK activates DAG
(see Figure 1). Note that each LIH has a unique underlying
interaction graph (which can be easily derived from the LIH
representation by splitting the AND connections), whereas the
opposite is, in general, not true.
Within this logical framework we may study the effect of a set of
input stimuli (typically ligands) on downstream signaling by comput-
ing the logical steady state [13] that results by propagating the signals
through the network from the input to the output layer. It seems
worthwhile to remark that the updating assumption (synchronous
versus asynchronous [14,15])—which must usually be made when
dealing with dynamic Boolean networks—is not relevant here as we
focus on the logical steady states, which are equivalent in both cases.
Sometimes a logical steady state is not unique or does not exist due to
the presence of feedbacks loops. However, many feedback loops
become active only in a longer timescale justifying setting them OFF
in the first wave of signal propagation (allowing them to be switched
ON for the second timescale). This has been used here for several
feedback loops (see main text and Table S2). The effect of knocking-
out a species can be tested by re-computing the (new) logical steady
state for the respective stimuli. MISs satisfying a given intervention
goal can be computed by systematically testing sets of permanently
activated or/and deactivated nodes [13,31].
All mathematical analyses and computations have been performed
with our software tool CellNetAnalyzer [31], a comprehensive user
interface for structural analysis of cellular networks. CellNetAnalyzer
and the T cell model can be downloaded for free (for academic use)
from http://www.mpi-magdeburg.mpg.de/projects/cna/cna.html.
Immunoblotting. Human or mouse T cells were purified using an
AutoMACS magnetic isolation system according to the manufac-
turer’s instructions (Miltenyi, http://www.miltenyibiotec.com). Mouse
T cells were stimulated with 10 lg/ml of biotinylated CD3e (a subunit
of the TCR) antibody (145–2C11, BD Biosciences, http://www.
bdbiosciences.com/), 10 lg/ml of biotinylated CD28 antibody (37.51,
BD Biosciences), CD3 plus CD28 mAbs, or with CD3 plus 10 lg/ml of
biotinylated CD4 (GK1.5, BD Biosciences) followed by crosslinking
with 25 lg/ml of streptavidin (Dianova, http://www.dianova.de) at 37
8C for the indicated periods of time. Human T cells were stimulated
with CD3e mAb MEM92 (IgM, kindly provided by Dr. V. Horejsi,
Prague, Czech Republic) or with CD3 plus CD28 mAbs (248.23.2).
Cells were lysed in buffer containing 1% NP-40, 1% laurylmaltoside
(N-dodecyl b-D-maltoside), 50 mM Tris pH 7.5, 140 mM NaCl, 10mM
EDTA, 10 mM NaF, 1 mM PMSF, 1 mM Na3VO4. Proteins were
separated by SDS/PAGE, transferred onto membranes, and blotted
with the following antibodies: anti-phosphotyrosine (4G10), anti-
ERK1/2 (pT202/pT204), anti-JNK (pT183/pY185), anti-phospho-Akt
(S473) (all from Cell Signaling, http://www.cellsignal.com/), anti-
ZAP70 (pTyr 319, Cell Signaling), anti-ZAP70 (cloneZ24820, Trans-
duction Laboratories, http://www.bdbiosciences.com/), or against b-
Actin (Sigma, http://www.sigmaaldrich.com/). Where PI3K and src-
kinase inhibitors were used, T cells were treated with 100 nM
Wortmannin (Calbiochem, http://www.emdbiosciences.com) or 10 lM
PP2 (Calbiochem) for 30 min at 37 8C prior to stimulation. All
experiments have been repeated three times and reproduced the
shown results.
Supporting Information
Table S1. List of Compounds in the Logical T Cell Model
Model name corresponds to the name in Figure 1 and Table S2.
Common abbreviations are those usually used in the literature, while
name is the whole name. Type classifies the molecules, if applies, as
follows: K ¼ Kinase, T ¼ Transcription Factor, P ¼ Phosphatase, A ¼
Adaptor Protein, R ¼ Receptor, G ¼ GTP-ase. In the case where two
pools of a molecule were considered, a ‘‘reservoir’’ was included
which was required for both pools. This allows us to perform a
simultaneous knock-out of both pools.
Found at doi:10.1371/journal.pcbi.0030163.st001 (56 KB PDF).
Table S2. Hyperarcs of the Logical T Cell Signaling Model (see Figure
1 and Methods)
Exclamation mark (‘‘!’’) denotes a logical NOT, and dots within the
equations indicate AND operations. The names of the substances in
the explanations are those used in the model and Figure 1; the
biological names are displayed in Table S1. In the case where two
pools of a molecule were considered (e.g., lckp1 and lckp2), a
‘‘reservoir’’ (lckr) was included which was required for both pools.
This allows us to perform a simultaneous knock-out of both pools
acting on the reservoir.
Found at doi:10.1371/journal.pcbi.0030163.st002 (183 KB PDF).
Acknowledgments
The authors would like to thank the members of the signaling group at
the Institute of Immunology (M. Smida, X. Wang, S. Kliche, R. Pusch, M.
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1589
A Logical Model of T Cell Receptor Signaling
Togni, A. Posevitz, V. Posevitz, T. Drewes, U. Ko¨ lsch, S. Engelmann) and
I. Merida and J. Huard for essential biological input into the model.
Author contributions. JSR set up the model and performed the
analysis. LS, JAL, UB, BA, and BS, with the help of the signaling group
at the Institute of Immunology, gathered the biological details of the
model and analyzed the correctness of the results. LS and BA
performed the wet-lab experiments. RH supported the model setup,
analysis, and documentation. SK developed the theoretical methods
and tools (CellNetAnalyzer) and supported the analysis. UUH and RW
contributed to the theoretical methods with useful insights. EDG, BS,
and RW coordinated the project.
Funding. The authors are thankful for the support of the German
Ministry of Research and Education to EDG (Hepatosys), the German
Research Society to BS and EDG (FOR521), and the Research Focus
Dynamical Systems funded by the Saxony-Anhalt Ministry of
Education.
Competing interests. The authors have declared that no competing
interests exist.
References
1.
Kitano H (2002) Computational systems biology. Nature 420: 206–210.
2.
Alon U, Surette MG, Barkai N, Leibler S (1999) Robustness in bacterial
chemotaxis. Nature 397: 168–171.
3.
Wiley HS, Shvartsman SY, Lauffenburger DA (2003) Computational
modeling of the EGF-receptor system: A paradigm for systems biology.
Trends Cell Biol 13: 43–50.
4.
Sasagawa S, Ozaki Y, Fujita K, Kuroda S (2005) Prediction and validation of
the distinct dynamics of transient and sustained ERK activation. Nat Cell
Biol 7: 365–373.
5.
Altan-Bonnet G, Germain RN (2005) Modeling T cell antigen discrim-
ination based on feedback control of digital ERK responses. PLoS Biol 3:
356.
6.
Papin JA, Hunter T, Palsson BO, Subramaniam S (2005) Reconstruction of
cellular signalling networks and analysis of their properties. Nat Rev Mol
Cell Biol 6: 99–111.
7.
Oda K, Matsuoka Y, Funahashi A, Kitano H (2005) A comprehensive
pathway map of epidermal growth factor receptor signaling. Mol Syst Biol.
doi:10.1038/msb4100014
8.
Jeong H, Mason SP, Barabasi AL, Oltvai ZN (2001) Lethality and centrality
in protein networks. Nature 411: 41–42.
9.
Milo R, Shen-Orr S, Itzkovitz S, Kashtan N, Chklovskii D, et al. (2002)
Network motifs: Simple building blocks of complex networks. Science 298:
824–827.
10. Ma’ayan A, Jenkins SL, Neves S, Hasseldine A, Grace E, et al. Formation of
regulatory patterns during signal propagation in a Mammalian cellular
network. Science 309: 1078–1083.
11. Stelling J, Klamt S, Bettenbrock K, Schuster S, Gilles ED (2002) Metabolic
network structure determines key aspects of functionality and regulation.
Nature 420: 190–193.
12. Price ND, Reed JL, Palsson BO (2004) Genome-scale models of microbial
cells: Evaluating the consequences of constraints. Nat Rev Microbiol 2: 886–
897.
13. Klamt S, Saez-Rodriguez J, Lindquist J, Simeoni L, Gilles ED (2006) A
methodology for the structural and functional analysis of signaling and
regulatory networks. BMC Bioinformatics 7: 56.
14. Kauffman SA (1969) Metabolic stability and epigenesis in randomly
constructed genetic nets. J Theor Biol 22: 437–467.
15. Thomas R, D’Ari R (1990) Biological feedback. Boca Raton (Florida): CRC
Press.
16. Mendoza L, Thieffry D, Alvarez-Buylla ER (1999) Genetic control of flower
morphogenesis in Arabidopsis thaliana: A logical analysis. Bioinformatics 15:
593–606.
17. Albert R, Othmer HG (2003) The topology of the regulatory interactions
predicts the expression pattern of the Drosophila segment polarity genes. J
Theor Biology 223: 1–18.
18. Chaves M, Albert R, Sontag ED (2005) Robustness and fragility of Boolean
models for genetic regulatory networks. J Theor Biol 235: 431–449.
19. Benjamini E, Coico R, Sunshine G (2000) Immunology—A short course.
New York: Wiley-Liss.
20. Huang Y, Wange RL (2004) T cell receptor signaling: Beyond complex
complexes. J Biol Chem 279: 28827–28830.
21. Lee KH, Holdorf AD, Dustin ML, Chan AC, Allen PM, et al. (2003) T cell
receptor signaling precedes immunological synapse formation. Science 295:
1539–1542.
22. Chan C, Stark J, George AT (2004) Feedback control of T-cell receptor
activation. Proc Biol Sci 271: 931–939.
23. Mendoza L (2006) A network model for the control of the differentiation
process in Th cells. Biosystems 84: 101–114.
24. Meng W, Sawasdikosol S, Burakoff SJ, Eck MJ (1999) Structure of the
amino-terminal domain of Cbl complexed to its binding site on ZAP-70
kinase. Nature 398: 84–90.
25. Duan L, Reddi AL, Ghosh A, Dimri M, Band H (2004) The Cbl family and
other ubiquitin ligases destructive forces in control of antigen receptor
signaling. Immunity 21: 7–17.
26. Thien CB, Langdon WY (2005) c-Cbl and Cbl-b ubiquitin ligases: Substrate
diversity and the negative regulation of signalling responses. Biochem J 15:
153–166.
27. Csete M, Doyle J (2004) Bow ties, metabolism and disease. Trends
Biotechnol 22: 44–450.
28. Reth M, Brummer T (2004) Feedback regulation of lymphocyte signalling.
Nat Rev Immunol 4: 269–277.
29. Cinquin O, Demongeot J (2002) Positive and negative feedback: Striking a
balance between necessary antagonists. J Theor Biol 216: 229–241.
30. Remy E, Ruet P (2006) On differentiation and homeostatic behaviours of
Boolean dynamical systems, Lect Notes Comput Sci 4230: 153–162.
31. Klamt S, Saez-Rodriguez J, Gilles ED (2007) Structural and functional
analysis of cellular networks with CellNetAnalyzer. BMC Systems Biology
1: 2.
32. Cai YC, Cefai D, Schneider H, Raab M, Nabavi N, et al. (1995) CD28pYMNM
mutations implicate phosphatidylinositol 3-kinase in CD86-CD28-medi-
ated costimulation. Immunity 3: 417–426.
33. Rudd CE, Raab M (2003) Independent CD28 signaling via VAV and SLP-76:
A model for in trans costimulation. Immunol Rev 192: 32–41.
34. Su B, Jacinto E, Kallunki T, Karin M, Ben-Neriah Y (1994) JNK is involved
in signal integration during costimulation of T lymphocytes. Cell 77: 727–
736.
35. Harlin H, Podack E, Boothby M, Alegre ML (2002) TCR-Independent CD30
signaling selectively induces IL-13 production via a TNF receptor-
associated factor/p38 mitogen-activated protein kinase-dependent mecha-
nism J Immunol 169: 2451–2459.
36. Gravestein LA, Amsen D, Boes M, Calvo CR, Kruisbeek AM, et al. (1998) The
TNF receptor family member CD27 signals to Jun N-terminal kinase via
Traf-2. Eur J Immunology 28: 2208–2216.
37. Hanke JH, Gardner JP, Dow RL, Changelian PS, Brissette WH, et al. (1996)
Discovery of a novel, potent, and Src family-selective tyrosine kinase
inhibitor. Study of Lck- and FynT-dependent T cell activation. J Biol Chem
271: 695–701.
38. Holdorf AD, Green JM, Levin SD, Denny MF, Straus DB, et al. (1999)
Proline residues in CD28 and the Src homology (SH)3 domain of Lck are
required for T cell costimulation. J Exp Med 190: 375–384.
39. Kovacs B, Parry RV, Ma Z, Fan E, Shivers DK, et al. (2005) Ligation of
CD28 by its natural ligand CD86 in the absence of TCR stimulation
induces lipid raft polarization in human CD4 T cells. J Immunol. 175:
7848–7854.
40. Tybulewicz VL (2005) Vav-family proteins in T-cell signalling. Curr Opin
Immunol 17: 267–274.
41. Arnold R, Patzak IM, Neuhaus B, Vancauwenbergh S, Veillette A, et al.
(2005) Activation of hematopoietic progenitor kinase 1 involves relocation,
autophosphorylation, and transphosphorylation by protein kinase D1. Mol
Cell Biol 25: 2364–2383.
42. Hehner SP, Hofmann TG, Dienz O, Droge W, Schmitz ML (2000) Tyrosine-
phosphorylated Vav1 as a point of integration for T-cell receptor- and
CD28-mediated activation of JNK, p38, and interleukin-2 transcription. J
Biol Chem 275: 18160–18171.
43. Herishanu Y, Kay S, Rogowski O, Pick M, Naparstek E, et al. (2005) T-cell
ZAP-70 overexpression in chronic lymphocytic leukemia (CLL) correlates
with CLL cell ZAP-70 levels, clinical stage and disease progression.
Leukemia 19: 1289–1291.
44. Chang F, Lee JT, Navolanic PM, Steelman LS, Shelton JG, et al. (2003)
Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and
neoplastic transformation: A target for cancer chemotherapy. Leukemia
17: 590–603.
45. Bentires-Alj M, Gil SG, Chan R, Wang ZC, Wang Y, et al. A role for the
scaffolding adapter GAB2 in breast cancer. Nat Med 12: 114–121.
46. Noh DY, Kang HS, Kim YC, Youn YK, Oh SK, et al. (1998) Expression of
phospholipase C-gamma 1 and its transcriptional regulators in breast
cancer tissues. Anticancer Res 18: 2643–2648.
47. Stelling J, Sauer U, Szallasi Z, Doyle FJ III, Doyle J (2004) Robustness of
cellular functions. Cell 118: 675–685.
48. Schrijver A (2003) Combinatorial optimization. Berlin: Springer.
49. Yablonski D, Kuhne MR, Kadlecek T, Weiss A (1998) Uncoupling of
nonreceptor tyrosine kinases from PLC-gamma1 in an SLP-76-deficient T
cell. Science 281: 413–416.
50. Sugie K, Jeon MS, Grey HM (2004) Activation of naive CD4 T cells by anti-
CD3 reveals an important role for Fyn in Lck-mediated signaling. Proc Natl
Acad Sci U S A 101: 14859–14864.
51. Schaeffer EM, Debnath J, Yap G, McVicar D, Liao XC, et al. (1999)
Requirement for Tec kinases Rlk and Itk in T cell receptor signaling and
immunity. Science 284: 638–641.
PLoS Computational Biology | www.ploscompbiol.org
August 2007 | Volume 3 | Issue 8 | e163
1590
A Logical Model of T Cell Receptor Signaling
|
17722974
|
tcrlig = ( tcrlig )
vav1 = ( zap70 AND ( ( ( sh3bp2 ) ) ) ) OR ( xx )
lat = ( zap70 )
pi3k = ( ( xx ) AND NOT ( cblb ) ) OR ( ( lckp2 ) AND NOT ( cblb ) )
gap = ( unknown_input3 )
rac1r = ( unknown_input )
Dummy = ( ( plcgb AND ( ( ( vav1 AND zap70 AND itk AND slp76 ) ) ) ) AND NOT ( ccblp2 ) )
ca = ( ip3 )
pkb = ( pdk1 )
nfkb = NOT ( ( ikb ) )
lckp1 = ( ( cd45 AND ( ( ( lckr AND cd4 ) AND ( ( ( NOT csk ) ) ) ) ) ) AND NOT ( shp1 ) )
nfat = ( calcin )
bclxl = NOT ( ( bad ) )
gadd45 = ( unknown_input )
bad = NOT ( ( pkb ) )
pag = ( ( fyn ) AND NOT ( tcrb ) )
ccblp2 = ( ccblr AND ( ( ( fyn ) ) ) )
cyc1 = NOT ( ( gsk3 ) )
vav3 = ( sh3bp2 )
sre = ( rac1p2 ) OR ( cdc42 )
ikkg = ( card11a AND ( ( ( pkcth ) ) ) )
calcin = ( ( ( ( cam ) AND NOT ( calpr1 ) ) AND NOT ( akap79 ) ) AND NOT ( cabin1 ) )
cabin1 = NOT ( ( camk4 ) )
grb2 = ( lat )
ship1 = ( unknown_input2 )
plcga = ( ( plcgb AND ( ( ( vav1 AND zap70 AND itk AND slp76 ) ) ) ) AND NOT ( ccblp2 ) )
sh3bp2 = ( zap70 AND ( ( ( lat ) ) ) )
shp2 = ( gab2 )
pten = ( unknown_input2 )
mlk3 = ( hpk1 ) OR ( rac1p1 )
gsk3 = NOT ( ( pkb ) )
cd45 = ( unknown_input )
dag = ( ( plcga ) AND NOT ( dgk ) )
bcl10 = ( unknown_input2 )
gab2 = ( grb2 AND ( ( ( lat AND zap70 ) ) ) ) OR ( gads AND ( ( ( lat AND zap70 ) ) ) )
erk = ( mek )
ap1 = ( fos AND ( ( ( jun ) ) ) )
fos = ( erk )
cblb = NOT ( ( cd28 ) )
gads = ( lat )
pdk1 = ( pip3 )
itk = ( pip3 AND ( ( ( zap70 AND slp76 ) ) ) )
ras = ( ( sos AND ( ( ( rasgrp ) ) ) ) AND NOT ( gap ) )
ikb = NOT ( ( ikkab ) )
ccblp1 = ( ccblr AND ( ( ( zap70 ) ) ) )
p21c = NOT ( ( pkb ) )
tcrp = ( tcrb AND ( ( ( fyn OR lckp1 ) ) ) )
rac1p2 = ( rac1r AND ( ( ( vav3 ) ) ) )
rsk = ( erk )
fyn = ( lckp1 AND ( ( ( cd45 ) ) ) ) OR ( tcrb AND ( ( ( lckr ) ) ) )
hpk1 = ( lat )
cam = ( ca )
cre = ( creb )
akap79 = ( unknown_input2 )
p70s = ( pdk1 )
p38 = ( ( zap70 ) AND NOT ( gadd45 ) )
card11a = ( malt1 AND ( ( ( card11 AND bcl10 ) ) ) )
pip3 = ( ( ( pi3k ) AND NOT ( ship1 ) ) AND NOT ( pten ) )
card11 = ( unknown_input )
ccblr = ( unknown_input )
creb = ( rsk )
zap70 = ( ( tcrp AND ( ( ( abl ) ) ) ) AND NOT ( ccblp1 ) )
jun = ( jnk )
camk2 = ( cam )
jnk = ( mkk4 ) OR ( mekk1 )
sos = ( grb2 )
slp76 = ( ( zap70 AND ( ( ( gads ) ) ) ) AND NOT ( gab2 ) )
mkk4 = ( mekk1 ) OR ( mlk3 )
cdc42 = ( unknown_input2 )
lckp2 = ( lckr AND ( ( ( tcrb ) ) ) )
mekk1 = ( hpk1 ) OR ( cdc42 ) OR ( rac1p2 )
lckr = ( lckr_input )
plcgb = ( lat )
ip3 = ( plcga )
raf = ( ras )
ikkab = ( ikkg AND ( ( ( camk2 ) ) ) )
rlk = ( lckp1 )
fkhr = NOT ( ( pkb ) )
cd28 = ( cd28 )
malt1 = ( unknown_input )
pkcth = ( dag AND ( ( ( pdk1 AND vav1 ) ) ) )
p27k = NOT ( ( pkb ) )
rasgrp = ( dag )
mek = ( raf )
rac1p1 = ( rac1r AND ( ( ( vav1 ) ) ) )
tcrb = ( ( tcrlig ) AND NOT ( ccblp1 ) )
abl = ( fyn ) OR ( lckp1 )
csk = ( pag )
xx = ( cd28 )
shp1 = ( ( lckp1 ) AND NOT ( erk ) )
dgk = ( tcrb )
bcat = NOT ( ( gsk3 ) )
calpr1 = ( unknown_input2 )
camk4 = ( cam )
|
BioMed Central
Page 1 of 26
(page number not for citation purposes)
BMC Bioinformatics
Open Access
Methodology article
A methodology for the structural and functional analysis of signaling
and regulatory networks
Steffen Klamt*†1, Julio Saez-Rodriguez†1, Jonathan A Lindquist2,
Luca Simeoni2 and Ernst D Gilles1
Address: 1Max-Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, D-39106 Magdeburg, Germany and 2Institute for
Immunology, University of Magdeburg, Leipziger Strasse 44, D-39120 Magdeburg, Germany
Email: Steffen Klamt* - klamt@mpi-magdeburg.mpg.de; Julio Saez-Rodriguez - saezr@mpi-magdeburg.mpg.de;
Jonathan A Lindquist - Jon.Lindquist@Medizin.Uni-Magdeburg.de; Luca Simeoni - Luca.Simeoni@Medizin.Uni-Magdeburg.de;
Ernst D Gilles - gilles@mpi-magdeburg.mpg.de
* Corresponding author †Equal contributors
Abstract
Background: Structural analysis of cellular interaction networks contributes to a deeper understanding
of network-wide interdependencies, causal relationships, and basic functional capabilities. While the
structural analysis of metabolic networks is a well-established field, similar methodologies have been
scarcely developed and applied to signaling and regulatory networks.
Results: We propose formalisms and methods, relying on adapted and partially newly introduced
approaches, which facilitate a structural analysis of signaling and regulatory networks with focus on
functional aspects. We use two different formalisms to represent and analyze interaction networks:
interaction graphs and (logical) interaction hypergraphs. We show that, in interaction graphs, the
determination of feedback cycles and of all the signaling paths between any pair of species is equivalent to
the computation of elementary modes known from metabolic networks. Knowledge on the set of signaling
paths and feedback loops facilitates the computation of intervention strategies and the classification of
compounds into activators, inhibitors, ambivalent factors, and non-affecting factors with respect to a
certain species. In some cases, qualitative effects induced by perturbations can be unambiguously predicted
from the network scheme. Interaction graphs however, are not able to capture AND relationships which
do frequently occur in interaction networks. The consequent logical concatenation of all the arcs pointing
into a species leads to Boolean networks. For a Boolean representation of cellular interaction networks
we propose a formalism based on logical (or signed) interaction hypergraphs, which facilitates in particular
a logical steady state analysis (LSSA). LSSA enables studies on the logical processing of signals and the
identification of optimal intervention points (targets) in cellular networks. LSSA also reveals network
regions whose parametrization and initial states are crucial for the dynamic behavior.
We have implemented these methods in our software tool CellNetAnalyzer (successor of FluxAnalyzer) and
illustrate their applicability using a logical model of T-Cell receptor signaling providing non-intuitive results
regarding feedback loops, essential elements, and (logical) signal processing upon different stimuli.
Conclusion: The methods and formalisms we propose herein are another step towards the
comprehensive functional analysis of cellular interaction networks. Their potential, shown on a realistic T-
cell signaling model, makes them a promising tool.
Published: 07 February 2006
BMC Bioinformatics2006, 7:56
doi:10.1186/1471-2105-7-56
Received: 28 July 2005
Accepted: 07 February 2006
This article is available from: http://www.biomedcentral.com/1471-2105/7/56
© 2006Klamt et al; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 2 of 26
(page number not for citation purposes)
Background
Evolution has equipped cells with exquisite signaling sys-
tems which allow them to sense their environment,
receive and process signals in a hierarchically organized
manner and to react accordingly [1]. The complexity of
the corresponding molecular machineries, in accordance
with the complicated tasks they have to perform, is over-
whelming. In the last few years, as a key element to the
growing popularity of systems biology, mathematical
tools have been applied to the analysis of signaling data
[2]. Ordinary differential equations relying on kinetic
descriptions of the underlying molecular interactions are
arguably the most used approach for modeling signaling
networks (e.g. [3-6]). A number of theoretical methods
have been devised and employed for the reconstruction
(reverse engineering) of signaling or, more generally,
interaction networks (which may represent signaling but
also other types or abstractions of cellular networks such
as genetic regulatory networks) based on perturbation
experiments [7]. The approaches rely on methods ranging
from Bayesian networks (e.g. [8]) to metabolic control
analysis [9,10].
Relatively few methods have been proposed so far for ana-
lyzing the structure of a given signaling (or any interac-
tion) network. This is somewhat surprising since
structural analysis of metabolic networks is a well-estab-
lished field and proved to be successful to recognize rela-
tionships between structure, function, and regulation of
metabolic networks [11]. Structural analysis will be partic-
ularly useful in large signaling networks, where a simple
visual inspection is not possible and at the same time the
construction of precise quantitative models is practically
infeasible due to the huge amount of required, but gener-
ally unknown, kinetic parameters and concentration val-
ues. However, the reconstruction of large signaling
networks is still in its first stages [2,12].
Structural or qualitative approaches that have been
employed for interaction networks include statistical
large-scale analyses in protein-protein networks (e.g.
[13]). These studies are important for examining statisti-
cal properties of the interaction graph and for understand-
ing its global organization but they provide relatively few
insights into the function of the network. Papin and co-
workers [14,15] were the first to adapt methods from the
constraint-based approach (frequently used for structural
analysis of metabolic networks [11]) to analyze stoichio-
metric models of signaling pathways. Recently, graph-the-
oretical descriptions of signaling networks have been
examined [16-18]. Finally, Boolean networks as discrete
approximations of quantitative models have been used
for logical analyses of small signaling networks e.g. [19].
However, the majority of studies relying on the Boolean
approach deal with genetic interaction networks, many of
which have a relatively small size (ca. 10 species; e.g.
[20,21]), however, recently more complicated networks
have also been investigated [22,23].
In this contribution, we propose formalisms for represent-
ing signaling and other interaction networks mathemati-
cally and present a collection of methods facilitating
structural analysis of the respective network models.
Rather than introducing completely new concepts, we will
systematize and adapt existing formalisms and methods,
often motivated from structural analyses of metabolic net-
works, towards a functional analysis of the structure of a
signaling network. Issues that can be addressed with the
proposed methods include:
• check of the plausibility and consistency of the network
structure
• identification of all or particular signaling pathways,
feedback loops and crosstalks
• network-wide functional interdependencies between
network elements
• identification of the different modes of (logical) input/
output behavior
• predicting responses (phenotypes) after changes in net-
work structure
• finding targets and intervention points in the network
for repressing or provoking a certain behavior or response
• analysis of structural network properties like redun-
dancy and robustness
Structural analysis is not based on quantitative and
dynamic properties and can thus only provide qualitative
answers. However, some insights into the dynamic prop-
erties can nevertheless often be obtained, because funda-
mental properties of the dynamic behavior are often
governed by the network structure [24]. While we will
focus on signaling networks, the methods can be easily
applied to any kind of interaction network, including
gene regulatory systems. Apart from a toy model, we will
exemplify our methods on a model of signaling pathways
in T-cells.
Results and discussion
Mass and signal flows in cellular interaction networks
The reader familiar to the structural analysis of stoichio-
metric networks may notice that, in the case of metabolic
networks, many of the issues in the task list of the previ-
ous section have been handled by the constraint-based
approach [11]. For example, the identification of func-
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 3 of 26
(page number not for citation purposes)
tional pathways and studying the input (substrates)/out-
put (products) behavior of stoichiometric reaction
networks is facilitated by elementary-modes analysis
[25,26]. Flux Balance Analysis is another related tech-
nique often used for phenotype predictions of metabolic
mutants [11,27]. Recently, the concept of minimal cut sets
has been introduced for identifying targets in metabolic
networks [28,29]. Therefore, it seems reasonable to apply
these methods to signaling networks. However, some fun-
damental differences in the way the network elements
interact may complicate a direct transfer:
(1) The constraint-based framework assumes steady-state,
while in signaling networks a transient behavior can often
be observed. (However, as will be discussed below, many
useful insights of signaling networks can be obtained
from using a static approach.)
(2) In stoichiometric networks, any arrow (reaction) lead-
ing from educts to products can be seen as an "activating"
(producing) connection for the products. Therefore,
employing stoichiometric framework it is difficult or only
indirectly possible to express an inhibitory action of a spe-
cies onto another.
(3) Probably the most significant difference is that the
edges (i.e. the connections between the species) in meta-
bolic networks carry flows of mass whereas edges in sign-
aling networks may carry mass and/or information
(signal) flow. Of course, at the molecular level, any inter-
action between species in the cell can be written as a stoi-
chiometric equation. However, whereas mass flow is
connected to a real consumption of participating com-
pounds, signal flow is usually characterized by a recycling
of certain species (e.g. enzymes) so that these species can
mediate the signal transfer continuously (until they are
degraded).
A typical example, namely the activation of a receptor
tyrosine kinase (Figure 1(a)) [30], illustrates the simulta-
neous occurrence of mass and signal flow. A ligand (Lig)
binds to the extracellular domain of a receptor (Rec) yield-
ing a receptor-ligand complex which can undergo further
changes (e.g. by autophosphorylation or/and dimeriza-
tion). We denote the outcome by RecLig*. This complex is
now able to phosphorylate another molecule (M).
Accordingly, M binds to RecLig* and becomes phosphor-
ylated (M-P) by the expense of ATP. At the end, M-P is
released, recycling also the activated receptor-ligand com-
plex RecLig*.
The first step in this scheme can be considered as a mass
flow. However, the cycle in which RecLig* phosphorylates
M, is a mass flow with respect to M and ATP, but a signal-
ing flow with respect to RecLig*, as the latter is indeed
required for driving this cycle but not consumed (because
recycled) in the overall stoichiometry.
In performing a structural analysis we are interested in
extracting signaling paths from the network scheme.
Therefore, it may seem reasonable to compute elementary
modes, which typically represent pathways in reaction
networks with mass flow [25]. A basic property of elemen-
tary modes is that the (relative) mass flow represented by
an elementary mode keeps the "internal" species in a bal-
anced state. Internal species (here: RecLig*, RecLig*-M,
RecLig*-M-P) are within the system's boundary, whereas
the external species (here: Rec, Lig, M, M-P, ADP, ATP) are
considered as pools which are balanced by processes lying
outside the system's boundaries. Computing the elemen-
tary modes from the respective stoichiometric model of
Figure 1(a) gives exactly one mode which reflects the dis-
cussed role of RecLig* as a kinase (Figure 1(b)): in its net
stoichiometry, this elementary mode converts the external
species M and ATP into M-P and ADP, whereas RecLig* is
recycled. Since RecLig* is neither consumed nor produced
in the overall process, the first step (building the receptor-
ligand complex) is not involved in this mode simply
because a continuous synthesis of RecLig* would lead to
an accumulation of this species, which is inconsistent
with the steady-state assumption of elementary modes.
Thus, the causal dependency of M-P from the availability
of Rec and Lig is not reflected by the mass flow concept of
elementary modes. Note that exactly the same conceptual
problem would arise when enzymes and enzyme synthe-
(a) Example of a typical signaling pathway where mass and
signal flow occur simultaneosly (Rec = receptor; Lig = ligand;
RecLig* = (active) receptor-ligand-complex; M = molecule;
M-P = phosphorylated molecule M)
Figure 1
(a) Example of a typical signaling pathway where mass and
signal flow occur simultaneosly (Rec = receptor; Lig = ligand;
RecLig* = (active) receptor-ligand-complex; M = molecule;
M-P = phosphorylated molecule M). (b) The (only) elemen-
tary mode in this example which follows when M, M-P, ADP,
ATP, Rec and Lig are considered as external (boundary) spe-
cies. The involved reactions are indicated by green, thick
arrows. In its net stoichiometry, this elementary mode con-
verts M and ATP into M-P and ADP, whereas RecLig* is recy-
cled in the overall process. Importantly, the mandatory
process of building the receptor-ligand-complex RecLig*
(hence, the causal dependeny of M-P from the availability of
Rec and Lig) is not reflected by this mode.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 4 of 26
(page number not for citation purposes)
sis would be considered explicitly in stoichiometric stud-
ies of metabolic networks.
The example demonstrates that we require a framework
with the ability to account for mass and signal flows. Han-
dling both mass and signal flows formally equivalent as
interactions could be a suitable approach. Interpreting Fig-
ure 1(a)) as a diagram of interactions we could redraw it
as depicted in Figure 2(a). The dashed arrow indicates that
RecLig* catalyzes the phosphorylation of M to M-P. If we
assume that ADP, ATP, and M are always present, we get
the simple chain shown in Figure 2(b) expressing that Rec
and Lig are required to obtain RecLig* (or to activate
RecLig*), and that RecLig* is required to get M-P. If we do
not further distinguish between the two types of arrows
and thus consider mass and signal flows as formally
equivalent, the causal connections between the species
would, nevertheless, still be captured correctly. This
abstract representation of different types of interactions
will thus be used herein.
The following two sections will deal first with interaction
graphs and later with the more general (logical) interac-
tion hypergraphs. The basic difference between these two
related approaches can be illustrated by how they deal
with a connection such as "Rec + Lig" in Figure 2(b). If we
interpret it as "Rec activates RecLig* and Lig activates
RecLig*" then the concept of interaction graphs is applica-
ble (discussed in the following section). However, it
would be more accurate to say that "Rec AND Lig are
required simultaneously for building RecLig*", and it is this
more refined approach that leads to the concept of inter-
action hypergraphs, which will be discussed in further
details later on.
Analyzing interaction graphs
Definition of interaction graphs
Interaction (or causal influence) graphs are frequently
used to show direct dependencies between species in sig-
naling, genetic, or protein-protein interaction networks.
The nodes in these graphs may represent, depending on
the network type and the level of abstraction, receptors,
ligands, effectors, kinases, genes, transcription factors,
metabolites, proteins, and other compounds, while each
edge describes a relation between two of these species. In
signaling and gene regulatory networks, two further char-
acteristics are usually specified for each edge: a direction
(which species influences which) and a sign ("+" or "- ",
depending on whether the influence is activating (level
increasing) or inhibiting (level decreasing)). Formally, we
represent a directed interaction or causal influence graph
as a signed directed graph G = (V, A), where V is the set of
vertices or nodes (species) and A the set of labeled
directed edges [31,32]. Directed edges are usually called
arcs and an arc from vertex i (tail) to j (head) is denoted by
an ordered tuple {i,j,s} with i, j ∈ V and s ∈ {+,- }.
Sometimes, for example in protein-protein interaction
networks, the directions of the edges remain unspecified.
We will not consider such undirected interaction graphs
explicitly, however, many of the issues discussed in the
following can be transferred to undirected graphs (e.g. by
representing an undirected edge by two (forward and
backward) arcs).
The structure of a signed graph can be stored conveniently
by an m x q incidence matrix B in which the columns cor-
respond to the q arcs (interactions) and the rows to the m
nodes (species), similar as in stoichiometric matrices of
metabolic reaction networks [33]. For the k-th arc {i, j, s}
a (-1) is stored in the k-th column of B for the tail vertex
(i) and (+1) for the head vertex (j) of arc k. Hence, Bi,k = -
1 and Bj,k = 1 and Bl,k = 0 (l≠ i, j). For storing the signs, a q-
vector s is introduced whose k-th element is (+1) if arc k is
positive and (-1) if k is negative.
Self-loops (arcs connecting a species with itself) are not
considered here but could be stored in a separate list since
they would appear as a zero column in the incidence
matrix.
Note that, as far as the memory requirement is concerned,
the structure of a graph can be stored more efficiently than
by an incidence matrix, e.g. by using adjacency lists [34].
However, since we will present methods directly operat-
ing on the incidence matrix, we refer herein to this repre-
sentation.
Signal transduction networks are usually characterized by
an input, intermediate, and output layer (cf. [16]). The
input domain consists only of species having no predeces-
Interpretation of Figure 1 as an interaction network
Figure 2
Interpretation of Figure 1 as an interaction network.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 5 of 26
(page number not for citation purposes)
sor, which can thus not be activated from other species in
the graph. Such sources (typical representatives are recep-
tors and ligands) are starting points of signal transduction
pathways and can easily be identified from the incidence
matrix since their corresponding row contains no positive
entry. In contrast, the output layer consists only of nodes
having no successor. These sinks, usually corresponding to
transcription factors or genes, are identifiable as rows in B
which have no negative entry. The set of source and sink
nodes define the boundaries of the network under inves-
tigation. They play here a similar role as the external
metabolites in stoichiometric studies [33]. The intermedi-
ate layer functions as the actual signal transduction and
processing unit. It consists of the intermediate species, all
of which have at least one predecessor and at least one
successor, i.e. they are influenced and they influence other
elements. Such species contain both -1 and +1 entries in
the incidence matrix. In reconstructed signaling networks,
the detection of all sink and source species may help to
detect gaps in the network, e.g. when a species should be
an intermediate but is classified as a sink or source.
The presence of sinks and sources are a consequence of
setting borders to the system of interest. Sometimes there
are no sinks or/and no sources, especially in models of
gene regulatory networks (see e.g. the networks studied in
[21]), but this does not impose limitations to the
approaches presented here.
A toy example of a (directed) interaction graph that will
serve for illustrations throughout this paper is given in
Figure 3. This interaction graph, called TOYNET, consists
of two sources (I1, I2), two sinks (O1, O2), 7 intermediate
species (A,..., G), two inhibiting (arcs 2 and 7) and 11 acti-
vating interactions. Incidence matrix B of TOYNET reads
(the sign vector s is given on the top of B):
Identification of feedback loops
Even though some analysis methods (e.g. Bayesian net-
works) rely on acyclic networks where feedbacks are not
allowed, one of the most important features of signaling
and regulatory networks are their feedback loops
[3,5,18,21,35-38]. Positive feedbacks are responsible and
even required [39] for multiple steady state behavior in
dynamical systems. In biological systems, multistationar-
ity plays a central role in differentiation processes and for
epigenetic and switch-like behavior. In contrast, negative
feedback loops are essential for homeostatic mechanisms
(i.e. for adjusting and maintaining levels of system varia-
bles) or for generating oscillatory behavior [35].
Most reports demonstrating the role and consequences of
feedback loops analyze relatively small networks where
the cycles can be easily recognized from the network
scheme but rather few works address the question of how
feedback cycles can be identified systematically. This is
particularly important in large interaction graphs, where a
detection by simple visual inspection is impossible, espe-
cially when feedback loops overlap.
A feedback loop is, in graph theory, a directed cycle or cir-
cuit. A circuit is defined as a sequence C = {a1,...,aw} of arcs
that starts and ends at the same vertex k and visits (with
the exception of k) no vertex twice, i.e. C = {a1,...,aw} =
{{k, l1}, {l1,l2},..., {lw-1,k}} such that all nodes k, l1, l2 ...
lw-1 are distinct. The parity of the number of negative signs
of the arcs in C determines whether the feedback loop is
negative (odd number of negative signs) or positive
(even). In the example TOYNET two feedback loops can
be found: (i) the arc sequence {4,5,6,7} which is negative
(since one negative arc (7) is involved), and (ii) the
sequence {10,11}, which is positive (because the signs of
both arcs in this circuit are positive). Obviously, sinks and
sources (and all arcs connected to these nodes) can never
be involved in any circuit.
B =
+
−
+
+
+
+
−
+
+
+
+
+
+
−
−
−
−
1
1
0
0
0
0
0
0
0
0
0
0
0
0
0
1
0
0
0
0
0
0
0
0
0
0
0
0
0
1
0
0
0
0
0
0
0
0
1
1
1
0
0
1
1
0
0
0
0
0
0
0
0
0
0
0
0
1
1
0
0
0
0
1
0
0
0
0
0
0
1
1
0
0
0
0
0
0
0
1
1
0
0
0
0
1
1
0
0
0
−
−
−
−
−
−
0
0
0
0
0
0
0
0
0
1
1
1
0
0
0
0
0
0
0
0
0
0
0
1
1
0
1
0
0
0
0
0
0
0
0
0
0
0
1
0
0
0
0
0
0
0
0
0
0
0
0
0
1
−
−
−
I1
I2
A
B
C
D
E
F
G
O1
O2
1( )
Example of a directed interaction graph (TOYNET)
Figure 3
Example of a directed interaction graph (TOYNET). Arcs 2
and 7 indicate inhibiting interactions, while all others are acti-
vating.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 6 of 26
(page number not for citation purposes)
Computing all directed cycles in large graphs is computa-
tionally a difficult task. Algorithms that can be found in
the literature usually rely on backtracking strategies (e.g.
[16,40]). Here, we introduce a different approach where
the circuits are identified as elementary modes establish-
ing a direct link to metabolic network analysis. Circuits
can be formally represented by a q-vector c in which ci = 1
if arc i is involved in the circuit and ci = 0 otherwise. A cir-
cuit vector fulfills the equation
B c = 0 (2)
and hence, lies in the null space of the incidence matrix of
the graph [32,41]. Generally, any vector c obeying (2) ful-
fills a so-called conservation law and is called a circulation
which may be envisioned as a flow cycling around in the
network [42]. Eq. (2) is strongly related to the mass bal-
ance equation of metabolic networks in steady state. In
fact, considering the graph as a reaction network with the
arcs being irreversible mono-molecular reactions, the inci-
dence matrix would be equivalent to the stoichiometric
matrix and any circulation would be equivalent to a sta-
tionary flux distribution. Note that not all circulations are
circuits: the linear combinations of circuit vectors do also
yield circulations but are not (elementary) circuits. Pre-
cisely, circuits are special circulations having two addi-
tional properties. First, they must be admissible with
respect to the directions of the involved arcs, i.e. only non-
negative values are allowed for c:
ci≥ 0 for all i. (3)
Second, circuits are non-decomposable circulations, i.e. the
set of arcs building up the circuit c, expressed by P(c) = {i:
ci > 0}, is irreducible:
There is no non-zero vector d fulfilling eqs. (2) and (3)
and P(d)⊂ P(c) (4)
Eqs. (2) and (3) and condition (4) close the complete
analogy to elementary modes. In fact, cycles or circuits are
the elementary modes in the special case of graphs (ele-
mentary modes are defined for any matrix in eq. (2), not
only for the very special shape of incidence matrices
related to graphs). Any feasible stationary flux vector in a
metabolic network can be obtained by non-negative lin-
ear combinations of elementary modes. Equivalently, any
circulation vector can be decomposed into a non-negative
linear combination of circuit vectors. Note that, multiply-
ing a (circuit) vector c, that fulfills (2)-(4), by a scalar b>0
yields another vector v = bc which represents the same cir-
cuit because the same arcs compose it (are unequal to
zero). Moreover, all non-zero components in a circuit vec-
tor are equal to each other. Therefore we can always nor-
malize the vector in such a way that we obtain the binary
representative of this circuit where all components are
either "1" or "0".
In metabolic networks, elementary modes reveal not only
internal cycles but also, even with higher relevance, meta-
bolic pathways connecting input and output species. Con-
tinuing with the analogy to interaction graphs, in the next
subsection we will see that elementary modes can be used
to identify not only feedback loops but also signaling
paths.
Signaling (influence) paths between two species
When the interaction graph is very large it becomes diffi-
cult to see whether a species S1 can influence (activate or
inhibit) another species S2 and via which distinct path-
ways this can happen. Computing the complete set of
directed paths between a given pair (S1, S2) of species is
therefore often desirable. A path P = {a1,...,aw} is, similarly
to a feedback circuit, a sequence of arcs where none of the
nodes is visited more than once, but in the case of a sign-
aling path the start node S1 is distinct from the end node
S2, i.e. P = {a1,...,aw} = {{S1,l1}, {l1,l2}, ..., {lw-1,S2}} such
that all nodes S1, S2, l1, l2 ... lw-1 are distinct.
To obtain the signaling pathways from S1 to S2 we pro-
ceed as follows (Figure 4(a)): we add an "input arc" for S1
(i.e. a new column in the incidence matrix B containing
only zeros except a (+1) for S1) and an "output arc" for S2
(another new column in B containing only zeros except a
(-1) for S2. Then, computation of the elementary modes
in this network will provide the original feedback loops
without participation of the input and the output arc (as
shown above) and additionally all paths starting with the
input arc at S1 and ending with the output arc at S2, with
the latter revealing all possible routes between S1 and S2.
Computation of all signaling paths between two species
(here: between I1 and O1)
Figure 4
Computation of all signaling paths between two species
(here: between I1 and O1). (a) via the incorporation of a
"simplified" input and output arc; (b) with explicit introduc-
tion of an ENV („environment") node. Computing the ele-
mentary modes from the respective incidence matrix for (a)
and (b) yields basically the same result, namely all paths
between I1 and O1, as well as the two feedback circuits in
the intermediate layer.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 7 of 26
(page number not for citation purposes)
Admittedly, the introduced input and output arcs have no
tail or no head, respectively, and would therefore not be
edges in the graph-theoretical sense, but this has no con-
sequence for the analysis described within this contribu-
tion. In fact, this procedure is equivalent to adding in the
incidence matrix a "dummy" node representing the envi-
ronment (ENV), an "input arc" from ENV to S1 and an
"output arc" from S2 to ENV (Figure 4(b)). Computing
the elementary modes from the resulting incidence matrix
would produce the feedback circuits as well as the circuits
running over ENV. The latter represent the paths leading
from S1 to S2. In the procedure described above ENV is
simply removed from the incidence matrix leading to the
same results.
In order to obtain only the paths from S1 to S2 (without
the feedback loops), one can enforce the input and output
arc to be involved by using an extension of the algorithm
for computing elementary modes [43].
Furthermore, we may also add several input and output
edges simultaneously. For example, if we are interested in
all the paths connecting the input layer with the output
layer, i.e. all routes leading from a source to a sink node,
we add to each source an input edge and to each sink an
output edge and compute the elementary modes (and,
optionally, discard the feedback circuits where neither a
source nor a sink participates). In this way we obtain the
same set of signaling paths as if the elementary modes
would be computed separately for each possible pair of
source and sink nodes. Figure 5 shows the complete set of
signaling paths connecting the input with the output layer
of TOYNET.
Analogously to the feedback loops, we assign to each sig-
naling path an "overall sign" indicating whether A acti-
vates (+) or inhibts (-) B along this path. Again, the parity
of the signs of the arcs in the path determine whether the
influence is positive (even number of negative signs) or
negative (odd number of signs).
To sum up, feedback loops and influence paths in interac-
tion graphs can be identified as elementary modes (or,
equivalently, as extreme rays of convex cones [44]) from
the respective incidence matrix. Similar conclusions have
recently been drawn by Xiong et al. [45], albeit the authors
computed paths only between sink and source nodes and
only within unsigned graphs (i.e. they did not consider
inhibitory effects). Feedback circuits were also not consid-
ered. Hence, here we extend and generalize those results.
The equivalence of signaling paths and loops to elemen-
tary modes allows one the advantage to use the highly
optimized algorithms for computing elementary modes
[43,44,46].
Combinatorial studies on signaling paths
The computation of all paths between a pair of species
helps us to recognize all the different ways in which a sig-
nal can propagate between two nodes. In metabolic path-
way analysis, a statistical or combinatorial analysis of the
participation and co-occurrences of reactions in elemen-
tary modes proved to be useful for obtaining system-wide
properties, such as the detection of essential reactions/
enzymes or correlated reaction sets (enzyme subsets)
[11,26,47].
In principle, similar features are of interest also for signal-
ing paths and feedback loops. However, two important
issues arise in interaction graphs that require a special
treatment. First, we have two different types of pathways,
positives and negatives. Owing to their opposite mean-
ings we often need to analyze them separately in statistical
assessments. Second, in metabolic networks we are partic-
ularly interested in the reactions (edges), because they cor-
respond to enzymes that are subject to regulatory
processes and can be knocked-out in experiments. In con-
trast, in interaction graphs we are usually more interested
in the nodes, since they are often knocked-out in experi-
ments or medical treatments, either via mutations, siRNA
or by specific inhibitors. An edge in signaling networks
represents mostly a direct interaction between a pair of
species and has therefore no mediator. In some cases, an
edge can directly be targeted by e.g. a mutation at the cor-
responding binding site of one of the two nodes species
All signaling paths linking the input layer (source species) with the output layer (sink species) in TOYNET
Figure 5
All signaling paths linking the input layer (source species) with the output layer (sink species) in TOYNET.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 8 of 26
(page number not for citation purposes)
involved. Here, we will focus on species participation,
albeit similar computations can be made for the edges.
As mentioned several times, in signaling networks we are
often interested in all the different ways by which a certain
transcription factor (or any other species from the output
layer) can be activated or inhibited by signals arriving the
input layer. For this purpose, we compute all signaling
paths leading from source nodes located in the input layer
down to a certain sink species s of interest. We denote the
set of all these paths by Is, which can be dissected in the
two disjoint subsets of activating and inhibiting paths: I =
∫
. Each source species i can then be classified into
one of the following four influence classes with respect to
s:
(1) activator of s (i is involved in at least one path of
and in no path of
)
(2) inhibitor of s (i is involved in at least one path of
and in no path of
)
(3) ambivalent factor for s (i is involved in at least one
(inhibiting) path of
and in at least one (activating)
path of
)
(4) without any influence on s (i is not involved in any
path of Is)
In TOYNET, we see from Figure 5 that I2 is a pure activator
and I1 an ambivalent factor for O1. With respect to O2, I1
is an inhibitor and I2 again an activator. The qualitative
response of s after perturbing the level of a non-affecting
species, or of an inhibitor or activator can be predicted
unambiguously (namely unchanged or decreasing or
increasing, respectively) as long as the network has no
negative feedback loop. Negative feedback loops limit
such qualitative predictions for activators (or inhibitors):
if there is any path from an activator (inhibitor) to s that
touches a negative feedback loop (i.e. at least one species
on the path is involved in a negative feedback) then the
resulting effect in perturbation experiments can not be
predicted uniquely (cf. [36]). This case occurs in TOYNET
for I2 with respect to O1: I2 is an activator of O1 but the
only connecting path (P5 in Figure 5) goes through spe-
cies C which participates in the negative feedback circuit.
Thus, although at least a transient increase in O1 can be
expected after up-regulating I2, we cannot exclude that the
negative feedback drives the level of O1 below its initial
level at a certain time point after increasing the level of I2.
We therefore call an activator (inhibitor) p of s a total acti-
vator (total inhibitor) of s if there is no path from p to a spe-
cies in a negative feedback circuit that is in turn connected
to s.
Positive feedbacks do not limit these qualitative up/
down-predictions because they cannot change the mono-
tone effect of the respective input signal, e.g. when
increasing the level of I2 in TOYNET we can expect an
increase in the level of O2 after some time.
To summarize, regarding the influence of a species p on
another species s we have 6 possible cases: total and non-
total activator, total and non-total inhibitor, ambivalent
factor and non-influencing species. Note that, by comput-
ing the connecting signaling paths, this classification pro-
cedure can be applied not only between a source and a
sink node but also between any pair of species, e.g.
between a source and an intermediate, an intermediate
and a sink, and two intermediates. In TOYNET, for exam-
ple, F is a total activator of O2 and has no influence on
O1, whereas D is an inhibitor but not a total one of O1
because it is connected to (even involved in) a negative
feedback circuit.
Additionally, as the complement of incoming paths, we
can also determine the paths starting in a certain species s
showing us which nodes and arcs are reachable from (and
influenceable by) s. As a further generalization, sets of
incoming and/or outgoing paths can also be defined not
only for a single species s but also for a set S of species.
This might be useful, for example, when we are interested
in all paths ending (starting) in a certain subset of the sink
(source) nodes.
Investigations of influence and signaling paths as pro-
posed above provide, apart from pair-pair relationships
(e.g. "a is a (total) activator of b" or "a has no influence on
b"), global properties (e.g. a is a (total) activator of all sink
species). Some other useful structural features and con-
straints can be detected by a statistical or combinatorial
analysis of certain path sets (partially, similar ideas have
been proposed by [14] for stoichiometric models of sign-
aling networks):
• Essential species (arcs): When focussing on a specific sig-
naling event, e.g. the activation of a certain species by sig-
nals from the input layer, we may identify essential
species (or arcs) with respect to this event. For example,
species E and arc 9 are essential for activating O2 but non-
essential for the activating paths leading to O1 in
TOYNET.
Is
+
Is
−
Is
+
Is
−
Is
−
Is
+
Is
−
Is
+
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 9 of 26
(page number not for citation purposes)
• Species (arc) participation: A more quantitative measure
can be obtained by giving percentages of all those activat-
ing and/or inhibiting pathways, in which the species or
arc is involved. One may only relate the relative participa-
tion to the paths where the respective species or arc is
involved or to the complete set of paths. For example, I2
is involved in 50% of all positive paths coming from the
input layer and activating O1, while I2 is involved in
100% of all paths activating O2 (but only 50% of the
paths coming from I2 lead to O2). Arc 9 is involved in one
activating and one inhibiting path leading to O2. Thus,
only 50% of the paths running over this arc are activating,
however, it is involved in all (100%) activating paths con-
necting sources with O2. Similar considerations can be
done regarding feedback loops: in TOYNET, species D and
A as well as arcs 6, 7 and 11 are not involved in paths con-
necting input with output layers and have thus a special
importance in establishing the negative (D, A, arcs 6 and
7) and positive (arc 11) feedback. (Note that a similar
measure for the importance of a species or arc is between-
ness centrality [48]. This importance measure is well-
known in graph theory and checks how many shortest
paths between pairs of nodes are running over the respec-
tive node or arc.)
• Redundancy: The total number of paths activating
(inhibiting) a species is a measure for the redundancy in
the system.
• Path length: The length distribution of signaling paths
provides a rough idea on the compactness of the network
[18].
• Crosstalk: Using our framework, crosstalk might be
defined as a place (node) where paths from different
source nodes cross each other for the first time. For exam-
ple, E is a crosstalk species in TOYNET (signals of I1 and
I2 cross) whereas F and G are not. In some cases, however,
crosstalk is a more complex phenomenon where different
nodes are involved. For example, at species C a path com-
ing from I1 via B and another path from I2 via E meet each
other. However, I1 and I2 have also met earlier in E and,
additionally, the action of I1 on C via B is already influ-
enced by I2 in species B since I2 can act on B via the path
visiting E, C, D and A.
Distance matrix and dependency matrix
Some applications presented in this section require
exhaustive enumerations of signaling paths becoming
computationally challenging in large networks. However,
in some cases we only want to know whether any activat-
ing and/or any inhibiting path between two nodes exists
or whether there is any positive or any negative feedback
circuit in which a certain species is involved. For such
"existence questions" we can often apply standard meth-
ods from graph theory. A very useful object is the distance
matrix D which can be obtained with low computational
demand by computing the shortest distances (shortest
path lengths) between each pair of species (e.g. Dijkstra's
algorithm [32]). D has dimension m × m and the element
Dij stores the length of the shortest path for traveling from
node i to node j, being Dij = ∞ if no paths exists between i
and j. The distance matrix shows immediately
• which elements can be influenced by species i (the i-th
row of D)
• which nodes can influence species i (i-th column of D)
• whether feedback circuits exist: if the distance Dii from a
node i back to itself is finite, then i is involved in at least
one feedback loop. Furthermore, if Dij and the transposed
element Dji are finite, Dij, Dji≠∞, then a feedback between
species i and j exists.
By an extension of the usual shortest path algorithm (not
shown), we may also compute separately a matrix Dpos for
the shortest positive paths and another Dneg for the shortest
negative paths. Table 1 shows the distance matrices Dpos
and Dneg from TOYNET.
Note that by taking the minimum values from Dpos and
Dneg, D can be obtained. Moreover, the two matrices Dpos
and Dneg, whose computation is reasonably possible in
very large networks, are sufficient to classify all species
into (total/non-total) activators, (total/non-total) inhibi-
tors, ambivalent factors, and non-influencing nodes with
respect to a certain compound y. The reason is that this
classification requires only knowledge on the existence of
positive and negative paths between species pairs and on
the existence of negative feedback loops. For example, a
species x is a total activator of y if (i) at least one positive
path from x to y exits (
≠ ∞) and if (ii) no negative
path from x to y exists (
= ∞) and if (iii) for any spe-
cies z that is influenced by x (Dx, z ≠ ∞) and connected to y
(Dz, y ≠ ∞) it holds, that z is not involved in a negative feed-
back (
= ∞).
For representing species dependencies in a compact man-
ner, we introduce the dependency matrix M, which shows
all the pair-wise dependencies, e.g. by using 6 different
colors (for the 6 possible cases). Thereby, the color of
matrix element Mxy indicates whether species x is a total/
non-total activator or a total/non-total inhibitor or an
ambivalent factor or a non-influencing node for species y.
Dx y
pos
,
Dx y
neg
,
Dz z
neg
,
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 10 of 26
(page number not for citation purposes)
Again, x = y is allowed, indicating feedbacks. Figure 6
shows the dependency matrix for TOYNET.
Although the distance and dependency matrices store a
wealth of structural information in a very condensed
manner, some applications still require a full enumera-
tion of all available signaling paths. One case is the sys-
tematic determination of minimal cut sets.
Minimal cut and intervention sets in interaction graphs
Searching for intervention strategies in signaling networks
is of high relevance in experimental and, in particular,
medical applications. Recently, the concept of minimal cut
sets has been introduced, which facilitates the identifica-
tion of efficient intervention strategies (cuts) and, at the
same time, the recognition of potential failure modes in a
given biochemical reaction network [28,29]. Basically, in
the most general version, a minimal cut set (MCS) is
defined as a minimal (irreducible, non-decomposable)
set of cuts (or failures) of edges or/and nodes that
represses a certain functionality or behavior in the system
[29]. For example, assume we want to prevent the activa-
tion of the sink node O1 in TOYNET. By removing nodes
{B, E} one can be sure that an activation of O1 by an
external stimulus becomes infeasible. The set {B, E}
would thus be a cut set for preventing the activation of
O1. Moreover, it is minimal since neither the removal of
only B nor the removal of only E can guarantee that the
"inhibition task" is achieved. Another minimal cut set
would be {C}. C is thus essential for activating O1, as
would be confirmed by participation analysis of all paths
activating O1. A general algorithmic scheme for a system-
atic enumeration of MCSs in stoichiometric networks was
given in [29]:
(i) Define a deletion task
(ii) Compute all minimal functional units (elementary
modes) and specify the set of target modes that have to be
attacked in order to achieve the deletion task
(iii) Compute the so-called minimal hitting sets of the tar-
get modes
We could proceed here in a similar way. First, a deletion
task specifying the goal of our intervention is defined. In
our example, the deletion task is "Prevent the activation of
Dependency matrix of TOYNET
Figure 6
Dependency matrix of TOYNET. The color of a matrix ele-
ment Mxy has the following meaning: (i) dark green: x is an
total activator of y; (ii) light green: x is a (non-total) activator
of y; (iii) dark red: x is a total inhibitor of y; (iv) light red: x is
a (non-total) inhibitor of y; (v) yellow: x is an ambivalent fac-
tor for y; (vi) black: x does not influence y;
Table 1: Shortest length of positive/negative paths in TOYNET (∞= no path exists). Values in the diagonal indicate whether the
respective element is involved in a positive/negative feedback loop. See also the dependency matrix in Figure 6.
I1
I2
A
B
C
D
E
F
G
O1
O2
I1
∞/∞
∞/∞
4/4
1/∞
2/2
3/3
∞/1
∞/2
∞/3
3/3
∞/4
I2
∞/∞
∞/∞
∞/4
∞/5
2/∞
3/∞
1/∞
2/∞
3/∞
3/∞
4/∞
A
∞/∞
∞/∞
∞/4
1/∞
2/∞
3/∞
∞/∞
∞/∞
∞/∞
3/∞
∞/∞
B
∞/∞
∞/∞
∞/3
∞/4
1/∞
2/∞
∞/∞
∞/∞
∞/∞
2/∞
∞/∞
C
∞/∞
∞/∞
∞/2
∞/3
∞/4
1/∞
∞/∞
∞/∞
∞/∞
1/∞
∞/∞
D
∞/∞
∞/∞
∞/1
∞/2
∞/3
∞/4
∞/∞
∞/∞
∞/∞
∞/4
∞/∞
E
∞/∞
∞/∞
∞/3
∞/4
1/∞
2/∞
∞/∞
1/∞
2/∞
2/∞
3/∞
F
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
2/∞
1/∞
∞/∞
2/∞
G
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
1/∞
2/∞
∞/∞
1/∞
O1
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
O2
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
∞/∞
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 11 of 26
(page number not for citation purposes)
O1 by any external input". Hence, the signaling paths
from the input layer to O1 are computed, which are P1,
P2, and P5 (see Figure 5). However, according to our dele-
tion task, the target set comprises only the paths P1 and
P5, because only these two activate O1. Finally, the mini-
mal hitting sets of the target paths have to be computed,
which are the MCSs [26,29]. When cutting species, a hit-
ting set T is a set of species that "hits" all target paths in a
minimal way, i.e. for each target path there is at least one
species that is contained in T and in the path. To be a min-
imal hitting set, no proper subset of T fulfills the hitting set
condition. The minimal hitting sets of the target paths and
hence the MCSs of our deletion task would be: {C}, {B,
E}, {I2, B}, {I1, E} and {I1, I2}. Deletion tasks may be
more complicated: for example, in TOYNET we might be
interested to repress the activation of O1 and O2. Accord-
ingly, the target paths would increase by one (P4 in Figure
5) resulting in another set of MCSs.
This example might suggest that we can use the same pro-
cedure as in metabolic networks, namely computing the
minimal hitting sets with respect to the target paths. This
naive approach works indeed for the case where the target
paths do only involve positive arcs (as in our example). It
can also be applied for interrupting any set of feedback cir-
cuits. For example, removing {A} interrupts the negative
feedback circuit and deleting {D, F} interrupts both feed-
back circuits in TOYNET. However, in general, negatively
signed arcs occurring in interaction graphs require a spe-
cial treatment. Even the following simple activating path
leading from a source species I to a sink species O contains
pitfalls:
. If the activation of O
is to be repressed, the signal flow along this path must be
interrupted. Removal of one species in the chain should
be sufficient. However, not all nodes are allowed to be cut.
If species B is removed, its negative action on C would be
interrupted, enabling in turn C to activate O. The reason
is that B, according to the definitions, is an inhibitor of O
and is therefore not a proper cut candidate. In fact, we
could add (constitutively provide or activate) B to stop an
activation of O. Generally, for attacking an activating path,
only the species that have an activating effect on the end
node of this path are proper cut candidates, whereas spe-
cies inhibiting the end node should instead be kept at a
high level to prevent an activation along this path. Hence,
as a generalization of (minimal) cut sets, we define (min-
imal) intervention sets (MISs) in interaction networks as
(minimal) sets of elements that are to be removed or to be
added in order to achieve a certain intervention task. By
allowing only the removal of elements, the set of MISs
coincides with the MCSs.
The computation of the MISs (or the smaller set of MCSs)
for a set of activating target paths that involve negatively
signed arcs is a more difficult task than computing only
minimal hitting sets. Indeed, each MIS will still represent
a hitting set, because at least one species in each target
path must be removed or constitutively provided. The dif-
ficulty arises by ambivalent factors which have in some
target paths an activating and in others an inhibitory effect
upon the end node. We could therefore restrict the inter-
ventions to those species that are either pure activators
with respect to the target paths (these are allowed to be
removed) or pure inhibitors (these are allowed to be
added). Using only these species, the MISs could again be
computed as the minimal hitting sets.
However, for computing MISs that may also act on ambiv-
alent factors, we present a more general algorithm (here
for a given set of activating target paths):
(1) In each target path, the involved nodes are labeled by
+1 (if the species influences the end node of the respective
path positively) or by -1 (if the species has a negative influ-
ence on the end node of the respective path).
(2) Combinations Ci of one, two, three, ... distinct
removed or activated species are constructed systemati-
cally. For each combination Ci, it is checked for each target
path whether the signal flow from the start node to the
end node is interrupted properly. A requirement is that at
least one of the positive (+1) species of each path is
removed or at least one negative (-1) species is provided
(added) by Ci (hitting set property). If, for a certain path,
Ci contains several nodes that are visited by this paths then
it is only checked whether the node closest to the end
node is attacked properly. When all paths have been
attacked (hit) properly by a combination Ci, then a new
MIS has been found. When constructing further combina-
tions of larger cardinality, the algorithm has to ensure that
none of the new combinations contains an earlier found
MISs completely.
Of course, this enumerative algorithm is even more time
consuming than computing minimal hitting sets and it
will become infeasible to compute all MISs in large net-
works. We may then restrict ourselves to MISs of low car-
dinality and/or to the subset of MCSs. Besides, the
determination of MISs can become even more compli-
cated: it might happen that a MIS attacks all activating tar-
get paths correctly but simultaneously destroys an
inhibiting path (not contained in the set of target paths)
which might then become an activating path. The MCS
{I1, I2} of our example represents such a problematic
I
A
B
C
O
+
−
−
+
→
→
→
→
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 12 of 26
(page number not for citation purposes)
case: it hits the two activating paths to O1 as demanded,
but it also attacks the inhibiting path leading from I1 to
O1. Thus, the inhibition of E through I1 would be inter-
rupted and it could be sufficient to retain E in an active
state enabling the activation of O1. Hence, we would not
be sure about the activation status of O1 after removing
this cut set. To avoid such side-effects, we may extend our
algorithm given above by checking also the consequence
of each intervention Ci with respect to the non-target
paths and exclude combinations that do not fulfill certain
criteria.
In a completely analogous fashion, we can also determine
MCSs or MISs that repress inhibitory paths. For example,
removing {I1} is a MCS that attacks the only inhibiting
path to O1, alternatively we might use the MISs {#E} or
{#C}, where # stands for "constitutively provided". The
same issues as discussed above must be taken into account
when interrupting a negative path: here, in each target
path, only the inhibiting species of the final sink source
should be removed whereas the activating nodes can be
added. Furthermore, we may also define more compli-
cated intervention tasks, e.g. where some activating and
some inhibiting paths are selected as target paths.
Jacobian matrix and interaction graph
Several works have highlighted the strong relationships
between interaction graphs and the Jacobian matrix J, the
latter obtained from a dynamical model of the network
under investigation [10,35,39]. A dynamic model of a sig-
naling (or any kind of interaction) network is usually
described by a system of ordinary differential equations
that model the evolution of the m network components x1
... xmwith the time:
The m × m Jacobian matrix J(x) collects the partial deriva-
tives of F with respect to x:
The sign of Jik(x) tells whether xk has a (direct) positive or
negative influence on xi and sign(J(x)) can thus be seen as
the adjacency matrix of the underlying interaction graph.
In an adjacency matrix Y, a non-zero entry for Yik indicates
an edge from node i to k. Adjacency and incidence matrix
are equivalent for describing a graph structure and can be
converted into each other: each non-zero element Yik gets
a corresponding column in the incidence matrix.
The sign structure of the Jacobian matrix is, in biological
systems, typically constant and reflects, despite its very
qualitative nature, fundamental properties of the dynamic
system. For example, multistationarity can only occur if a
positive circuit exists in the associated interaction graph
[39]. Methods for the detection of multistability in a spe-
cial class of dynamical systems – monotone I/O systems –
have been developed by Sontag et al. [36]. Monotone I/O
systems possess a monotonicity property that can be
checked from the interaction graph spanned by the Jaco-
bian matrix. In fact, having one source species and one
sink species, the required monotonicity property is equiv-
alent to our definition of a total activator of the sink node.
Thus, the methods developed in the previous section may
support such studies, where the structure of the Jacobian
matrix is analyzed. Having the absolute values of the Jaco-
bian matrix available (which change over time), arcs,
paths, and feedback circuits could be assigned an interac-
tion strength useful to identify key elements in the net-
work.
Boolean networks and (logical) interaction hypergraphs
Definitions
The methods described above consider an interaction as a
dependency between two species allowing to employ
tools from graph theory. However, in cellular networks,
an interaction (edge) often represents a relationship
among more than two species (nodes). A typical example
is a bimolecular reaction of the form A+B→ C, where three
species are involved. The binding of the ligand to the
receptor in Figure 2(b) (Rec+Lig→ RecLig*) is such a
bimolecular interaction. Using an interaction graph, this
reaction is modeled with two arcs (Figure 7(a)), namely
Rec→ RecLig* and Lig→ RecLig*, capturing correctly that
Rec and Lig have an influence on RecLig*. However, this
relaxed representation has shortcomings for a functional
interpretation of the network. To exemplify this, consider
the minimal cut sets repressing the phosphorylation of M
in Figure 7(a). As explained in the previous section, we
need to attack all positive paths leading to M-P. There are
two positive paths, one starting from Rec and the other
from Lig and, thus, {Rec, Lig} would be a minimal cut set.
But, intuitively, this cut set is not minimal for the real sys-
tem because both Rec and Lig are required for activating
M, and removing only one of the two species is thus suffi-
cient to interrupt the activation of M. (In other words, the
existence of a signaling path in an interaction graph does
not ensure that a signal can flow along this path.)
This example reveals that a proper consideration of AND-
connections between species is required. However, AND-
relationships are not possible in graphs but in hyper-
#
#
x
x
x
x
x
=
=
=
d
x
x
t
f
t
m
m
dt
t =
f
1
F( , )
1( , )
( , )
5
( )
J
x
x
ik
i
k
df
dx
( )
( )
=
( )
6
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 13 of 26
(page number not for citation purposes)
graphs, which are generalizations of graphs. Similar to a
directed graph, a directed hypergraph H=(V, A) consists of
a set V of nodes and a set A of hyperarcs (= directed hyper-
edges [49]). A hyperarc aconnects two subsets of nodes: a
= {S,E}; S,E⊂ V. S comprises the tail (start) nodes and E
the head (end) nodes of the connection. S and E can have
arbitrary cardinality, and a graph is a special case of a
hypergraph where the cardinality of S and E is 1 for all
edges.
In our context, without loss of generality, we will usually
have only one end node in E and we interpret a hyperarc
as an interaction in which the compound contained in E
is activated by a combined action of the species contained
in S. Figure 7(b) depicts the example with the receptor-lig-
and-complex as a hypergraph in which a hyperarc cap-
tures now the AND-connection between Rec and Lig
yielding RecLig*.
AND connections facilitate a refined representation of sto-
ichiometric conversions within interaction networks,
albeit the precise stoichiometric coefficients are not cap-
tured here. Apart from stoichiometric interactions, AND
connections allow the description of other dependencies,
for example, the case where only the presence of an acti-
vator AND the absence of an inhibitor leads to the activa-
tion of a certain protein.
In TOYNET, the four nodes (B, C, E, F) have more than
one incoming arc (Figure 3). In these nodes it is undeter-
mined how the different stimuli are combined, e.g.
whether B AND E are required to activate C or whether
one of both is sufficient (B OR E).
We could therefore concatenate all incoming edges in a
node by logical operations leading to Boolean networks
[21,31]. An assumption underlying Boolean networks is
to consider only discrete (concentration/activation) levels
for each species; in the simplest case a species can only be
"off" (= 0 = "inactive" or "absent") and "on" (= 1 =
"active" or "present"). Hence, each species is considered
as a binary (logical) variable. Next, a Boolean function fi
is defined for each node i which determines under which
conditions i is on or off, respectively. fi depends only on
those nodes in the interaction graph from which an arc
points into species i. In general, for constructing a Boolean
function, all logical operations like AND, OR, NOT, XOR,
NAND can be used. However, here we express each
Boolean function by a special representation known as
sum of products (SOP; also called (minimal) disjunctive
normal form (DNF)) which is possible for any Boolean
function [50]. SOP representations require only AND, OR
and NOT operators. In a SOP expression, literals, which
are Boolean variables or negated Boolean variables, are
connected by AND's giving clauses. Several such AND
clauses are then in turn connected by OR's. Using the
usual symbols '·' for AND, '+' for OR and '!' for NOT, an
example of a SOP expression would be: fi = x·y·z + x·!z
stating that fi gets value "1" if (x AND y AND z are active)
OR (if x is active AND z is NOT) and "0" else. The SOP
expression fi = x·!y + !x·y mimics an XOR gate.
In our context, writing a Boolean function as a SOP has
several advantages. First, many biological mechanisms
that lead to the activation of a species correspond directly
to SOP representations. Second, by using SOPs, the struc-
ture of a Boolean network can be represented and
depicted intuitively as a hypergraph: each hyperarc point-
ing into a node i is an AND clause of other nodes and rep-
resents one way of activating i; hence, all hyperarcs ending
in i are OR'ed together. A hyperarc carries a signal flow to
its end node and the binary value of the flow depends on
the state of all its start nodes. In the following, such a
hypergraph induced by a minimal SOP representation of
a Boolean network will be called a logical interaction hyper-
graph (LIH).
In Figure 8 a possible instance of a LIH compatible with
the interaction graph of TOYNET in Figure 3 is depicted.
In each of the four nodes with more than one incoming
arc, the logical concatenation has now been specified. For
example, B is now activated if A AND I1 are active simul-
taneously (hyperarc "1&4"). In contrast, C is activated if B
OR E is present (active), and F is active if E OR G are in an
active state. Hence, C and F retain their graph-like struc-
ture.
(a) the graphical and (b) the more correct hypergraphical
representation of the simple interaction network shown in
Figure 1 and 2
Figure 7
(a) the graphical and (b) the more correct hypergraphical
representation of the simple interaction network shown in
Figure 1 and 2.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 14 of 26
(page number not for citation purposes)
Inhibiting arcs in the interaction graph are interpreted in
the corresponding LIH as NOT-operations. Thus, arc 7 is
now interpreted as "A is active if D is not present". Since
arc 2 and 3 in Figure 3 have been combined with an AND
in Figure 8, we interpret this new hyperarc as "E becomes
activated if I2 is present AND I1 NOT". Hence, in contrast
to inhibiting arcs in interaction graphs, in general we do
not assign a minus sign (a NOT) to the complete hyperarc,
but to its negative branches (see hyperarc 2&3 in Figure
8), whereas all other branches get positive signs. Due to
the assignment of signs LIHs can formally be seen as
signed directed hypergraphs.
The pure logical description of a signaling or regulatory
network works well when the activation (inhibition) of a
species by others follows a sigmoid curve [21]. Problems
that might arise while describing a real network within the
logical framework and possible solutions are discussed in
a later section.
LIHs can be formally represented and stored in a similar
way as interaction graphs. The underlying hypergraph is
stored by an m × n incidence matrix B in which the rows
correspond to the species and the columns to the n hyper-
arcs. If species i is contained in the set of start (tail) nodes
of a hyperarc k then Bik = -1, if i is the endpoint (head) of
hyperarc k then Bik = 1, and if i is not involved in k we have
Bik = 0. For storing the NOTs operating on certain species
in a hyperarc we may use another m × n matrix U that
stores in Uik a "1" if species i enters the hyperarc k with its
negated value and "0" else. Accordingly, the incidence
matrix B for the LIH of TOYNET (Figure 8) reads
To be concise, the two non-zeros entries of U are indicated
by an asterisk in the incidence matrix.
Representing a Boolean network as a LIH we can easily
reconstruct the underlying interaction graph from the
matrices B and U: we simply split up the hyperarcs having
more than one start node (or/and more than one end
node in the general case). Thus, a hyperarc with d start and
g end nodes is converted into d·g arcs in the interaction
graph. The sign of each arc in the graph model can be
obtained from U. The reverse, the reconstruction of the
LIH from the interaction graph, is not possible in a unique
manner underlining the non-deterministic nature of
interaction graphs.
Time in Boolean networks
A logical interaction hypergraph describes only the static
structure of a Boolean network. However, it is the dynamic
behavior of Boolean networks that has been analyzed
intensely in the context of biological (especially genetic)
systems [21,31,51]. For studying the evolution of a logical
system we need to introduce the (discrete) time variable t
and a state vector x(t) that captures the logical values of
the m species at time point t. Two fundamental strategies
exist to derive the new state vector x(t+1) from the current
state x(t). In the synchronous model, the logical value of
each node i is updated by evaluating its Boolean function
fi with the current state vector: xi (t+1) = fi(x(t)). Synchro-
nous models are deterministic but assume for all interac-
tions the same time delay which is often too unrealistic for
biological systems [21]. In the asynchronous model, we
select any (but only one) node i whose current state is
unequal to its associated Boolean function: xi (t) ≠ fi(x(t)).
Only this node switches in the next iteration. Since there
are, in general, degrees of freedom in choosing the switch-
ing node, this description is non-deterministic. The
advantage is that the complete spectrum of potential tra-
jectories is captured, albeit the graph of sequences is usu-
ally very dense, complicating its analysis in large systems.
The asynchronous description becomes (more) determin-
istic if time delays for activation and inhibition events are
known [21].
B =
−
−
−
−
1
4 2
3
5
6
7
8
9
10
11
12
13
0
0
0
&
&
1
1(*)
0
0
0
0
0
0
0
0
0
0
1
0
0
0
0
0
0
0
0
0
1
0
1
0
0
0
0
0
0
1
0
0
0
0
0
0
0
0
0
0
1
0
1
0
0
0
0
0
0
1
0
0
0
0
0
0
0
1
0
0
0
0
0
0
0
0
1
1
0
1
1
1
1
−
−
−
−
−
−
(*)
0
0
0
0
0
1
1
0
0
0
0
0
0
0
0
1
0
1
0
0
0
0
0
0
0
0
0
1
0
0
0
0
0
0
0
0
0
0
0
1
−
−
−
1
0
1
I1
I2
A
B
C
D
E
F
G
O1
O2
( )
7
Logical interaction hypergraph of TOYNET (compare with
interaction graph in Figure 3)
Figure 8
Logical interaction hypergraph of TOYNET (compare with
interaction graph in Figure 3).
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 15 of 26
(page number not for citation purposes)
We are now approaching the main part of this section.
Logical steady-state analysis
An important characteristic of the dynamic behavior of
Boolean networks, which is equivalent for both asynchro-
nous and synchronous descriptions, is the set of logical
steady states (LSSs). LSSs are state vectors xs obeying
=
fi(xs) for all nodes i. Hence, in LSS, the state of each node
is consistent with the value of its associated Boolean func-
tion and, therefore, once a Boolean network has moved
into a logical steady state, it will stop to switch and then
retain this state.
In the following, we will focus on logical steady state anal-
ysis (thus circumventing any interpretation problems that
might arise by choosing synchronous or asynchronous
description), which suffices for a number of applications,
especially for predicting potential functional states in sig-
naling or regulatory networks.
Given a Boolean network we may enumerate all possible
LSSs [52]. However, this is computationally difficult in
large networks. Besides, we are often interested in particu-
lar LSSs that can be reached from a given initial state x0. In
some cases, we only know a fraction of all initial node val-
ues. For example, a typical scenario in signaling networks
would be that initial values from species in the input layer
are known (specifying which external signals reach the
cell and which not), and we would like to know how the
(logical) integration and propagation of these input sig-
nals generate a certain logical pattern in the output layer.
Of course, we have to "wait" until the signals reach the
bottom of the network and, for obtaining a unique
answer, there should be a time point from which the states
will not change in the future. This is equivalent to deter-
mining the LSS in which the network will run from a given
starting point.
In a possible scenario for TOYNET, the initial values of the
source species I1 and I2 might be known to be
= 0 and
= 1, whereas the initial states of all other nodes are
unknown (Figure 9(a)). The states of I1 and I2 will not
change anymore because I1 and I2 have no predecessor in
the hypergraph model. Assuming that each interaction
has a finite time delay, E must become active and B inac-
tive. From these fixed values we can conclude that C and
F will definitely become active (by E) at a certain time
point and not change this state in the future. Proceeding
further in the same way, we can resolve the complete LSS
resulting from the given initial values of I1 and I2 (Figure
9(b)).
The last example illustrated that partial knowledge on ini-
tial values, especially from the source nodes, can be suffi-
cient to determine the resulting LSS uniquely. However, in
general, several LSSs might result from a given set of initial
values or a LSS may not exist at all. For example, if we only
know
= 1 in TOYNET nothing can be concluded
regarding a LSS (except that I2 will retain its state). If no
complete LSS can be concluded uniquely from initial val-
ues, there might nevertheless be a subset of nodes that will
reach a state in which they will remain for the future. For
example, setting
= 1 E will definitely become inacti-
vated after some time (again, finite time delay is
assumed). Since in this scenario nothing further can be
derived for other nodes, we would say that xI1 = 1 and xE =
0 are partial LSSs for the initial value set {
= 1}. Note
that these two partial steady states would not change
when we specified more or even all initial values.
We have conceived an algorithm which derives partial
LSSs that follow from a given set of initial values (if for
each node a partial LSS can be found, then a unique and
complete LSS exists for the set of initial values). The itera-
tive algorithm uses the following rules in the logical
hypergraph model:
• initial values of source nodes will not change in the
future, hence, are partial LSSs
• if species i has a proved partial LSS of 0, all hyperarcs in
which i is involved with its non-negated value have a zero
flow
• if species i has a proved partial LSS of 1, all hyperarcs in
which i is involved with its negated value have a zero flow
xi
s
xI1
0
xI2
0
xI2
0
xI1
0
xI1
0
Example of a logical steady state in TOYNET resulting from a
particular set of initial states in the input layer
Figure 9
Example of a logical steady state in TOYNET resulting from a
particular set of initial states in the input layer.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 16 of 26
(page number not for citation purposes)
• if all hyperarcs pointing into node i have a zero flow,
then i has a partial LSS of 0
• if all start nodes of a hyperarc have a partial LSS of 1 (or
of 0 for those start nodes entering the hyperarc with the
negated value) then a partial LSS of 1 follows for the end
node of this hyperarc
• knowing all the positive feedback circuits in the system,
we can check whether there is a "self-sustaining" positive
circuit where the known initial state values of the involved
nodes guarantee a partial LSS for all the nodes in this cycle
(see comments below)
In each loop, the algorithm tries to identify new partial
LSSs (following from the current set of partial LSSs already
identified) until no further ones can be found. Setting ini-
tial values in the input layer, this can be envisioned as a
propagation of signals through the interaction network
until signals reach nodes where the available information
is not sufficient to derive a unique LSSs.
Generally, in logical interaction hypergraphs where the
underlying interaction graph has no feedback loop (i.e. is
acyclic), specification of the initial values of all the source
nodes will always result in a unique and complete LSS
since the signals can be propagated step by step from top-
down to the output layer. In general, if all initial values are
known for the input layer, non-uniqueness or even non-
existence of partial LSSs can only be generated by feedback
loops. The partial LSSs of nodes involved in positive feed-
backs do often depend on the initial values of all the
nodes in this loop. For example, defining
= 0 we can
conclude a partial LSSs of zero for E in TOYNET (Figure
8), but, among others, the values of F, G and O2 remain
unknown although the only connection to a source node
leads via E. The reason is that F and G build up a positive
feedback loop which cannot be resolved without knowl-
edge on further initial values. If we know, additionally to
= 0, that
= 1 then F and G will always keep
each other activated so that we can infer a partial LSS of 1
for F, G and O2 (this is the last rule in the list given
above). If we have instead
= 0, we derive a 0 for
the partial LSS of these three nodes. If one of the two
nodes F and G has an initial value of 1 and the other 0,
nothing can be derived since the positive loop might
become fully activated or fully deactivated. However,
what can be confirmed in these simple examples is that
positive feedback loops induce multistationarity. It is
noteworthy that continuous dynamic models of networks
with positive feedbacks will depend, apart from kinetic
parameters, in a similar fashion on initial state values.
In contrast, negative feedback loops are not sensitive
against initial values but they can be the source of oscilla-
tions, preventing hence the existence of LSSs. In TOYNET
we have one negative feedback loop which can potentially
generate oscillations, for example, when we set
= 1.
Then, C cannot be activated via E. Assuming an initial
value of 0 for C (the same conclusion would be drawn
with 1), D becomes deactivated and thus A actived. Due
to the partial LSS of 1 for I1 we get an activation of B and
then of C and D which in turn inhibits A leading in the
next round to a deactivation of B, C and D and so on. The
logical states within this circuit and downstream of it
(O1) will thus never reach a steady state. As shown in
[21], oscillatory behavior in logical models corresponds
to oscillations or a stable equilibrium (lying somehow
between the fully activated and fully inactivated level) in
the associated continuous model, depending on the cho-
sen parameters. Negative feedback loops can thus impede
predictions on the basis of logical steady states, but they
also point to network structures whose parametrization
will have great impact on the dynamic behavior.
Note that feedback loops do not always prevent predic-
tions on (partial) LSSs as can be seen by the example in
Figure 9, it depends on the given initial values.
Such a logical steady state or "signal flow" analysis (SFA)
as presented herein shares similarities with the established
method of metabolic flux analysis [53]. In MFA, uptake
and excretion rates of cells are measured in order to recon-
struct the intracellular flux distribution within a metabolic
network. MFA relies on the quasi-steady state assumption,
similarly as SFA relies on LSS. However, whereas MFA tries
to reconstruct the reaction rates along the edges and noth-
ing can be said on the states of the species, the goal of SFA
is to determine the steady states of the nodes (belonging
to a given activation scheme) from which then the signal
flows along the edges follow. It is noteworthy that the cal-
culability of unknown reaction rates in MFA depends only
on the set of known rates [54], whereas in SFA the set of
given initial states and their respective values determine the
unique calculability of (partial) LSSs.
Applications of logical steady state analysis
The LSS analysis introduced herein offers a number of
applications for studying functional aspects in cellular
interaction networks:
xI2
0
xI2
0
x
x
F
G
0
0
=
x
x
F
G
0
0
=
xI1
0
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 17 of 26
(page number not for citation purposes)
Input-output behavior
Imposing different patterns of signals in the input layer
one may check which species become activated or inhib-
ited in the intermediate and, in particular, in the output
layer. This can also be simulated in combination with dif-
ferent initial state values for certain intermediate nodes,
albeit this will have an influence on the LSS only in con-
nection with positive feedbacks, as shown above.
Mutants and interventions
The changes in signals flows and in the input-output
behavior occurring in a manipulated or malfunctioning
network can be studied by removing or adding elements
or by fixing the states of certain species in the network. In
TOYNET, for example, if we want to study the effect of a
mutant missing F (or the effect of adding an inhibitor for
F) we may remove species F from the network (or, equiv-
alently, fix the state of F to zero) and compute then the
partial LSSs again. We will see that, independently of a
given pattern in the input layer, G and O1 will be assigned
a partial LSS of 0. Removing elements often changes not
only the values, but also the determinacy of partial LSSs.
Minimal cut sets (MCSs) and minimal intervention sets (MISs)
The definiton of MCSs and MISs in logical interaction
hypergraphs is similar as in interaction graphs: a MCS is a
minimal (irreducible) set of species whose removal will
prevent a certain response or functionality as defined by
an intervention goal. In the more general MISs we permit,
additionally to cuts, also the constitutive activation of cer-
tain compounds. Two examples in TOYNET: removing F
is a MCS for repressing an activation of G and O2. Assum-
ing an initial state of zero for the species in the intermedi-
ate layer, adding I1 and removing B would be a proper
MIS for repressing the activation of O1 and O2. Note that
in the interaction graph of TOYNET, this intervention
would not suffice to attack all activating paths leading
from the input layer to O1 and O2 (path P4 not attacked,
Figure 5). This example underscores again that MCSs and
MISs in interaction hypergraphs are usually smaller than
those obtained from the underlying interaction graph,
simply because more constraints are added by logical
combinations. However, the determination of MCSs, and
let alone MISs, in logical interaction hypergraphs is com-
binatorially complicated as in interaction graphs, in par-
ticular when negative signs (NOTs) occur. Here, we can
only propose a "brute-force" approach where the LSS
analysis serves algorithmically as an oracle: we check sys-
tematically for each combination of one, two, three ...
knocked out (for MISs also of permanently activated)
nodes in the network how this affects the (partial) LSSs,
possibly in combination with a given scenario of initial
states. From the resulting partial LSSs we can decide
whether our intervention goal has been achieved or not.
To compute only minimal cut or intervention sets, further
combinations with a cut or intervention set already satis-
fying our intervention goal have to be avoided. The algo-
rithm can be stopped when a user-given maximum
cardinality for the MCSs/MISs has been reached.
Backward propagation
The methods described above compute partial LSSs actu-
ally only by forward propagation of signals, but one may
also do the opposite, e.g. fixing values in the output layer
and tracing back the required states of nodes in the inter-
mediate and input layer using similar rules as for forward
propagation.
Network expansion methods
There is an interesting relationship between our LSS anal-
ysis and network expansion methods proposed by Eben-
höh et al. [55]. Network expansion allows for checking
which metabolites can in principle be produced from a
provided set of start species within a metabolic (stoichio-
metric) reaction network. This is a special case in our log-
ical framework. Briefly, metabolic networks are per se
hypergraphs and can thus be represented as a LIH by using
only AND's (each reaction is an AND clause of its reac-
tants; stoichiometric coefficients are not considered) and
OR's. Hence, no inhibiting interactions exist. We may
then put the supplied set of available species in the input
layer, set the initial values of all other species to zero and
compute then the LSS. Note that, according to the expla-
nations given above, a complete LSS will always be found
since all initial values are given and no negative feedback
circuit exists. Therefore, the computed LSS indicates
which species can be produced from the input set and
which not.
Extensions for the logical description of interaction networks
Several extensions and refinements of the logical frame-
work can be introduced which allow a more appropriate
description of real signaling and regulatory networks:
(1) As already proposed and applied by Thomas et al.
[21], the discretization in more than two levels is in prin-
ciple possible. This mimics the fact, that in reality multi-
ple relevant threshold values for a species may exist. A
refined discretization could be relevant, for instance, for a
species that activates/inhibits more than one species (with
different threshold levels). Another relevant situation
occurs if a species can be activated via two paths (con-
nected by an OR; see species C in TOYNET): the activation
via both paths might be significantly stronger than by only
one. However, considering several activation levels for a
certain species forces one to often consider multiple levels
for elements downstream or/and upstream of this species,
increasing hence the complexity of the network, and
requiring detailed knowledge which is often not available.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 18 of 26
(page number not for citation purposes)
(2) As we have seen, negative feedback can limit the pre-
dictability in LSS analysis. However, in cellular networks,
negative feedbacks become activated often upon a certain
time period after an activation event occurs, for example,
when gene expression is involved. This might be consid-
ered by classifying species and/or hyperedges by assigning
a discrete time constant (or time scale) τ to each element
telling us whether this network element appears in an
early (τ = 1) or late (τ = 2) state. Using the sub-network
with all elements having a time constant of τ = 1 for the
first simulation and then using the computed LSSs as ini-
tial values for computing the second round (where the
complete network is considered) leads often to more real-
istic results. As in the case of multiple levels, this extension
requires a more detailed knowledge about the network
under consideration. An example in TOYNET (Figure 8):
we may assume that D is a factor that is transcriptionally
regulated by C, thus, arc 6 has a time constant of τ = 2 and
all others have τ = 1. Setting the initial values I1 = 1, I2 =
0 and D = 0 and computing the LSSs for τ = 1 activation of
C and O1 occurs. We can then fix the state of D (D = 1)
and get then a complete deactivation of C and O1.
(3) In real signaling and regulatory networks, it is some-
times difficult to decide whether arcs from the interaction
graph have to be linked by an AND or an OR in the inter-
action hypergraph. For example, in TOYNET, species E is
inhibited by factor I1 and activated by factor I2. If I1 has
a very strong inhibiting effect on E we may formulate the
hyperarc as done in Figure 8, suggesting that I1 must not
be active for activating E. However, if the interaction
strength of both I1 and I2 with respect to E is at the same
level (i.e. additive) neither "NOT(I1) OR I2" nor
"NOT(I1) AND I2" would reflect the real situation.
Indeed, this is a recurring situation in signaling networks,
where often a balance between different signals deter-
mines the activation of a certain element. At this point it
could be helpful to use logical operations that have a par-
tially incomplete truth table. In the latter example we
could say that E is active if (NOT(I1) AND I2) and E is
inactive if (I1 AND NOT(I2)). For the other two possible
cases, no decision could be made along this hyperedge. Of
course, modeling uncertainty in this way will limit the
determinacy but on the other hand a determined result
with this model allows a safer interpretation.
Analyzing interaction networks using CellNetAnalyzer
We have integrated many of the methods and algorithms
described herein in our software tool CellNetAnalyzer,
which is a MATLAB package and the successor of FluxAna-
lyzer [56]. Whereas FluxAnalyzer was originally developed
for structural and functional analysis of metabolic net-
works, CellNetAnalyzer extends these capabilities conse-
quently to the structural analysis of signaling and
regulatory networks. Apart from stoichiometric (meta-
bolic) reaction networks, CellNetAnalyzer supports now
also the composition of logical interaction hypergraphs
using AND, OR and NOT connections. Whenever needed,
the underlying interaction graph can be deduced from the
interaction hypergraph. Alternatively, by using only OR's
and NOT's, arbitrary interaction graphs can be con-
structed. As in FluxAnalyzer, the network model can be
linked with externally created graphics visualizing the net-
work. User interfaces (text boxes) enable data input and
output directly in these interactive maps (see screenshot
in Figure 10). New functions for graph-theoretical and
logical analysis have been integrated into the user menu;
Screenshot of the CellNetAnalyzer model for T-cell activation
Figure 10
Screenshot of the CellNetAnalyzer model for T-cell activation.
Each arrow finishing on a species box represents a hyperarc
and all the hyperarcs pointing into a species box are OR con-
nected. In the shown "early-event" scenario, the feedbacks
were switched off whereas all input arcs are active. The
resulting logical steady state was then computed. Text boxes
display the signal flows along the hyperarcs (green boxes:
fixed values prior computation; blue boxes: hyperarcs acti-
vating a species (signal flow is 1); red boxes: hyperarcs which
are not active (signal flow is 0)).
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 19 of 26
(page number not for citation purposes)
the results from computations are directly displayed
within the interaction maps or in separate windows. The
functions include:
• large-scale computation of all (positive and negative)
signaling paths connecting inputs with outputs or of all
signaling paths between a given pair of nodes; statistical
analysis of these paths
• large-scale computation of all (positive and negative)
feedback loops; statistical analysis of these routes
• computation of minimal cut sets for a given set of paths
or/and loops
• computation of distance (shortest paths) matrices – sep-
arately for positive and negative paths
• large-scale dependency analysis: identification of (total)
activators, (total) inhibitors and ambivalent factors for a
given species; display of the dependency matrix
• computation of (partial) logical steady states from a
given set of initial state values
• computation of (logical) minimal cut sets repressing or
provoking a user-defined behavior in the logical network
To illustrate the ability of our approach to deal with real
complex signaling networks, we have set-up and analyzed
in CellNetAnalyzer a logical model of T-cell activation (Fig-
ure 10), which will be discussed in the next section.
CellNetAnalyzer is free for academic purposes (see web-site
[57]).
Logical model of T-cell activation
T-cell activation and the molecular mechanisms behind
T-lymphocytes play a key role within the immune system:
Cytotoxic, CD8+, T-cells destroy cells infected by viruses or
malignant cells, and CD4+ helper T-cells coordinate the
functions of other cells of the immune system, such as B-
lymphocytes and monocytes [58]. Loss or dysfunction,
especially of CD4+ T-cells (as it occurs e.g. in the course of
HIV infection or in immuno-deficiencies) has severe con-
sequences for the organism and results in susceptibility to
viral and fungal infections as well as in the development
of malignancies. The importance of T-cells for immune
homeostasis is due to their ability to specifically recognize
foreign, potentially dangerous, agents and, subsequently,
to initiate a specific immune response that is aimed at
eliminating them. T-cells detect foreign antigens by means
of their T-Cell Receptor (TCR) which recognizes peptides
only when presented on MHC (Major Histocompatibility
Complex) molecules. The peptides that are recognized by
the TCR are typically derived from foreign (e.g. bacterial,
viral) proteins and are generated by proteolytic cleavage
within so called antigen presenting cells (APCs). Subse-
quent to their production the peptides are loaded onto the
MHC-molecules and the assembled peptide/MHC-com-
plex is then transported to the cell surface of the APC were
it can be recognized by T-cells. The whole process of anti-
gen uptake, proteolytic cleavage, peptide loading onto
MHC, transport of the peptide/MHC complex to the sur-
face of the APC and the recognition of the peptide/MHC-
complex by the TCR is called antigen presentation and
provides the molecular basis for the fine specificity of the
adaptive immune response.
The binding of peptide/MHC to the TCR, and the addi-
tional binding of a different region of the MHC molecules
to so called co-receptors (CD4 in the case of helper T-cells
and CD8 in the case of cytotoxic T-cells), initiates a pleth-
ora of signaling cascades within the T-cell. As a result, sev-
eral transcription factors – most importantly, AP1, NFAT
and NFκB – are activated. These transcription factors, in
turn, control the cell's fate, e.g. whether it becomes acti-
vated and proliferates [59] or not.
In the following, a logical model describing some of the
main steps involved in the activation of CD4+ helper T-
cells (also applicable for CD8+ cytotoxic T-cells) will be
briefly introduced and analyzed (see Figure 10 and Table
2). Several players, in particular, some whose role and
activation is not completely understood, are not included
in our model and thus their effects are not considered or
lumped with others. Additionally, in several, currently
still controversial cases, we have assumed one of the pos-
sible hypotheses; however, this does not mean that we
propose this to be the correct description of the TCR-
induced signaling network; we just want to demonstrate
the applicability of our approach on a realistic, complex
case. It is out of the scope of this paper to analyze the com-
plete, highly-complex signaling machinery of a T-cell.
Here, the biochemical steps included in the signaling
pathway will be described briefly; for a detailed descrip-
tion we refer the reader to reviews such as [59,60] and the
references therein:
• Upon binding of peptide/MHC to the TCR, the first
main step in the TCR-mediated signaling cascade is the
activation of the Src-family protein tyrosine kinase p56lck
(in the following termed Lck), although the exact mecha-
nism is still unclear. We have included one well accepted
mechanism [61], which probably plays a major role but
may be combined with others (cf. Figure 10):
In resting T-cells, the major negative regulator of Lck,
the protein tyrosine kinase Csk (C-terminal Src-kinase) is
¾
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 20 of 26
(page number not for citation purposes)
bound via a SH2-domain to the constitutively tyrosine
phosphorylated transmembrane adaptor protein PAG
(Protein Associated with Glycosphingolipid enriched
microdomains) and consequently inhibits membrane-
bound Lck by phosphorylating a C-terminal negative reg-
ulatory tyrosine residue of the Src kinase.
Upon ligand binding, PAG is dephosphorylated by a so
far unknown protein tyrosine phosphatase, thereby lead-
ing to the detachment of Csk from PAG, and hence releas-
ing Lck from the inhibitory effect of Csk. The release of
Csk from PAG, together with the activity of the membrane
associated tyrosine phosphatase CD45 (which dephos-
phorylates Lck on the same inhibitory residue that is
phosphorylated by Csk), and the concomitant binding of
the MHC molecule to the coreceptor CD4, leads to full
activation of Lck (see Figure 10).
However, both CD4 and the TCR can also be stimulated
individually, e.g. by using monoclonal antibodies specifi-
cally directed at either of the molecules or using cell lines
expressing mutated forms of CD4 that cannot bind MHC
or cannot transmit signals.
A regulation of the enzymatic activity of CD45 is not
included in the model (basically because it is not yet clear
how CD45 is regulated in vivo), but, since CD45 is an
important regulatory element for T-cells, it is included as
an input signal, allowing the analysis of its effect and the
performance
of
CD45
knock-out
experiments.
After a few minutes, PAG is rephosphorylated [62],
probably by the Src-kinase Fyn, and subsequently Csk is
re-recruited to PAG inhibiting Lck again.
• Activated Lck can phosphorylate another member of the
Src-protein kinases, p59fyn, in the following termed Fyn
(Fyn can probably also be activated in a Lck-independent,
TCR-dependent manner [63]). Additionally, Lck phos-
phorylates the so called ITAMs (Immunoreceptor Tyro-
sine-based Activation Motifs) that are present in the
cytoplasmic domains of the TCR-complex (the latter if the
TCR is close to Lck, i.e., if there is a concurrent activation
of the TCR). Subsequently, the Syk-family protein tyrosine
kinase ZAP70 (Zeta Associated Phosphoprotein of 70
kDa) binds to the phosphorylated ITAMs and, if Lck is
active, becomes activated by Lck-mediated tyrosine phos-
phorylation. Thus, during the initial phase of signal trans-
duction via the TCR three tyrosine kinases become
activated in a sequential manner, first Lck and Fyn and
then ZAP70. Together these three kinases propagate the
TCR-mediated signal by phosphorylating a number of
membrane associated and cytosolic signaling proteins.
• Active ZAP70 can phosphorylate LAT (Linker for Activa-
tion of T-cells), a second transmembrane adapter protein,
at four different tyrosine residues. Subsequently, cytoplas-
mic signaling molecules containing SH2-domains,
¾
¾
¾
¾
Table 2: The hyperarcs of the logical T-cell signaling model (see
Figure 10). Exclamation mark ('!') denotes a logical NOT and
dots within the equations indicate AND operations.
→ CD45
→ CD8
→ TCRlig
AP1 →
Ca → Calcin
Calcin → NFAT
CRE →
CREB → CRE
DAG → PKCth
ERK → Fos
ERK → Rsk
Fyn → PAGCsk
Fyn → TCRphos
Gads → SLP76
Grb2Sos → Ras
!IkB → NFkB
!IKKbeta → IkB
IP3 → Ca
JNK → Jun
Jun·Fos → AP1
LAT → Gads
LAT → Grb2Sos
LAT → PLCgbind
Lck·CD45 → Fyn
Lck → Rlk
MEK → ERK
NFAT →
NFkB →
!PAGCsk·CD8·CD45 → Lck
PKCth·DAG → RasGRP1
PKCth → IKKbeta
PKCth → SEK
PLCg(act) → DAG
PLCg(act) → IP3
Raf → MEK
Ras → Raf
RasGRP1 → Ras
Rsk → CREB
SEK → JNK
TCRbind·CD45 → Fyn
TCRbind·Lck → TCRphos
!TCRbind → PAGCsk
TCRlig·!cCbl → TCRbind
TCRphos·Lck·!cCbl → ZAP70
ZAP70·SLP76·PLCg(bind)·Itk → PLCg(act)
ZAP70·SLP76 → Itk
ZAP70 → cCbl
ZAP70 → LAT
ZAP70·SLP76·Rlk·PLCg(bind) → PLCg(act)
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 21 of 26
(page number not for citation purposes)
including the scaffolding proteins Grb2, Gads, and the
lipid kinase PLCγ1 (Phospholipase gamma 1), can bind to
phosphorylated LAT. Additionally, Grb2 binds to the
nucleotide exchange factor Sos (here we lumped Grb2 and
Sos in one activation step), and Gads to the adapter pro-
tein SLP76. The latter, upon phosphorylation by ZAP70,
can bind to the Tec-family tyrosine kinase Itk. Binding to
SLP76 and additional phosphorylation by ZAP70 acti-
vates Itk.
• For the activation of PLCγ1, the following conditions
have to be fulfilled: PLCγ1 is bound to LAT, SLP76 bound
to Gads, ZAP70 is activated (which hence phosphorylates
SLP76, allowing PLCγ1 to bind to SLP76), and Itk is
active, and hence is able to phosphorylate and thereby to
fully activate PLCγ1. Since all these conditions are needed,
a logical AND was included in the model (see Figure 10).
Rlk, another Lck-dependent Tec-family tyrosine kinase,
can also phosphorylate PLCγ1, hence Rlk has a redundant
role to Itk with regard to the activation of PLCγ1 [64].
• Activated PLCγ1 hydrolyses phosphatidyl-inositol-4,5
biphosphate (PIP2), which is considered an ubiquitous
membrane associated phospholipid and is therefore not
modeled, thereby generating the second messenger mole-
cules diacyloglycerol (DAG) and inositol trisphosphate
(IP3) [59,61].
• IP3 mediates calcium flux. Calcium (together with cal-
modulin) activates the serine phosphatase calcineurin,
which dephosphorylates the cytosolic form of the tran-
scription factor NFAT (Nuclear Factor of Activated T-
cells). The calcineurin-mediated removal of phosphate
groups allows NFAT to translocate to the nucleus and to
regulate gene expression.
• The second messenger DAG activates PKCθ and
(together with PKCθ[65]) activates the nucleotide
exchange factor RasGRP1.
• RasGRP1 and Sos (the latter if it is close to the mem-
brane, that is, if it is bound to LAT by means of Grb2), can
activate Ras, which in turn activates the Raf/MEK/ERK
MAPK Cascade.
• PKCθ is involved in the activation of JNK, as well as the
essential transcription factor NFκB (via phosphorylation
and subsequent degradation of the NFκB inhibitor, Iκ B,
by the PKCθ-activated Iκ B-kinase, IKK).
• ERK, activated by the Ras/Raf/MEK cascade, activates the
transcription factor CRE and (together with JNK) the
essential transcription factor AP1.
• The E3 ubiquitin ligase cCbl is important for shutting off
TCR-mediated signaling processes by ubiquitination of
key proteins, which are subsequently targeted for degrada-
tion [66]. One important target of cCbl is ZAP70; upon
tyrosine phosphorylation of ZAP70, cCbl binds to ZAP70,
leading to ZAP70's ubiquitination and degradation as
well as to the downregulation of the TCR.
From these biological facts we constructed a logical hyper-
graph model, containing 40 nodes and 49 hyperarcs, and
implemented it in CellNetAnalyzer (Figure 10). The model
is summarized in Table 2.
Remarks on the logical T-cell activation model
Note that a species can represent different states of a mol-
ecule: for example, CD45 refers to the availability of
CD45 to act on its substrates (Lck and Fyn), PLCg(bind)
refers to PLCγ1 bound to LAT, and PLCg(act) to the active
(bound to LAT and phosphorylated) form of PLCγ1. It is
also important to realize that several steps can be lumped
together or expressed in higher detail; for example, the
formation of the complex LAT:Grb2:Sos is considered as
one step, but intermediate steps could be considered. This
would be reasonable, for example, if Grb2 would have
other functions apart from binding Sos. Similarly, the two
steps of cCbl's effect (ubiquitination and degradation) are
lumped in the hyperarcs pointing to its targets ZAP70 and
TCR.
Also note that some of the logical operators could be
modeled in a different manner, as in the case of Sos and
RasGRP for the activation of Ras (where we prefer an OR
since both can independently activate Ras, although both
(AND) may be needed for full Ras activation).
Furthermore, our model describes the full activation of
the cascade which leads to proliferation; it is known that
e.g. stimulation of TCR with antibodies against its CD3
subunits produces a certain activation of the cascade
(where probably Fyn overtakes Lck's role [63]) but does
not lead to full activation. Therefore, in our model, as an
approximation, activated Fyn can phosphorylate the
ITAMs of the TCR, but is not able to activate ZAP70. Here
a model with more than 2 levels could be envisioned,
where activation of Fyn would be enough to produce a
weak (level 1) activation of ZAP70 and hence the whole
cascade downstream, while full activation via Lck would
activate the cascade to a level 2 (full activation).
The model has two extracellular input signals (one for the
TCR and one for the coreceptor CD4). Additionally, an
input arc for CD45 is included because the regulation of
CD45 is not modeled, as described above. Therefore,
mathematically speaking, the model contains 3 elements
in the input layer. On the other hand, the output layer
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 22 of 26
(page number not for citation purposes)
contains 4 transcription factors (CRE, AP1, NFAT and
NFκB).
As explained in the theoretical section, one reasonable
way to deal with the effect of negative feedbacks is to con-
sider the different time scales of the processes. Hence,
since PAG rephosphorylation takes place after a few min-
utes [62], and cCbl mediated degradation is an even
slower process, we can define several scenarios:
-τ = 0, resting-state (no inputs, no feedbacks),
-τ = 1, early-events (input(s), no feedbacks), and
-τ = 2, mid-time events (input(s), feedbacks). Here, the
state of the feedback loops (activation of PAG/Csk by Fyn
and recruitment of cCbl to phosphorylated ZAP70) will
depend on the state of the respective activators at τ = 1.
This can be considered either by fixing manually the state
values of cCbl and PAG/Csk for τ = 2 upon inspection at τ
= 1 (as was done herein) or by inclusion of a positive self-
loop.
We use the term mid-time event since one can also envi-
sion a long-term scenario (τ = 3), where slow gene expres-
sion mechanisms (not considered here) are active.
Analysis of the T-cell signaling cascade
In the interaction graph underlying the hypergraphical
model, there are 1158 paths from the input to the output
layer and 9 (7 negative and 2 positive) feedbacks loops,
which are listed in Table 3. cCbl is involved in most
(88%) of the loops, in accordance to its important role in
the regulation of the signaling cascade. Not surprisingly,
since the only feedback mechanisms included are the
effect of cCbl on ZAP70 and TCR and of Fyn on PagCsk,
no loop goes downstream of ZAP70, and a suitable mini-
mal cut set attacking all the feedback loops would consist
of Fyn and cCbl.
We further analyze the interaction graph by computing
the dependency matrix (Figure 11). Since downstream of
ZAP70 there are only positive connections (except at node
IκB), all the elements downstream of ZAP70 are total acti-
vators (except of IκB, which is a total inhibitor of NfκB)
with respect to the transcription factors in the output
layer, that is, they can have only positive effects. Therefore,
for these species, a negative intervention via e.g. inhibitors
or iRNA would unambiguously lead to a decrease in the
activation levels of the transcription factors. For consider-
ing the early-events scenario (τ = 1: the feedback loops are
not active), we recompute the dependency matrix where
the action of Fyn on PAGCsk and of ZAP70 on cCbl is not
considered (Figure 12). Then, all inputs (CD45, TCRlig
and CD4) are total activators for all species in the output
layer. This is not the case when the feedbacks become
active (Figure 11): TCRlig and CD45 become then ambiv-
alent factors, i.e. have negative connections to the sink
species, whereas CD4 is still an activator but no longer a
total one, as it is now connected to a negative feedback
loop.
A further analysis of the interaction graph provides that
there is no minimal cut set containing only one (essential)
species whose removal would interrupt all the positive
paths to all the outputs. In fact, all minimal cut sets satis-
fying this intervention task would contain at least two spe-
cies, for example MCS1 = {Rlk, ZAP70} and MCS2 =
{LAT, PLCg(act)}. The latter examples agree only partially
with biological knowledge: removal of MCS1 or MCS2
would indeed prevent the activation of any output, how-
ever, from experimental observations one knows that for
example LAT alone is essential in TCR signaling [60].
Thus, MCS2 would not be minimal.
Interpreting the hypergraphical (logical) model (Figure 10)
reveals that, due to several AND connections, the addi-
tional removal of PLCg(act) would indeed be redundant
because PLCg can anyway not be activated if LAT is
removed. This example illustrates the limitations of
graph-based methods and we computed therefore the
Table 3: All negative and positive feedback loops in the T-cell model as determined by CellNetAnalyzer. Negative influences are
indicated by "", positive influences are expressed by "→".
1 (negative)
TCRbind → TCRphos → ZAP70 → cCbl TCRbind
2 (negative)
TCRbind → Fyn → TCRphos → ZAP70 → cCbl TCRbind
3 (negative)
TCRbind PAGCsk Lck → ZAP70 → cCbl TCRbind
4 (negative)
TCRbind PAGCsk Lck → TCRphos → ZAP70 → cCbl TCRbind
5 (negative)
PAGCsk Lck → Fyn → PAGCsk
6 (negative)
TCRbind PAGCsk Lck → Fyn → TCRphos → ZAP70 → cCbl
TCRbind
7 (negative)
cCbl ZAP70 → cCbl
8 (positive)
TCRbind → Fyn → PAGCsk Lck → TCRphos → ZAP70 → cCbl
TCRbind
9 (positive)
TCRbind → Fyn → PAGCsk Lck → ZAP70 → cCbl TCRbind
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 23 of 26
(page number not for citation purposes)
(logical) minimal cut sets from the logical interaction
hypergraph revealing that not only LAT, but also ZAP70,
Lck, TCR, the ligand for the TCR, TCRphosp, CD4 and
CD45 are essential for full T-cell activation. This result is
in good agreement with the current knowledge: the T-cell
receptor, its ligand, and the ability of the receptor to get
phosphorylated are required for T-cell activation; and
CD4 (since it binds Lck thus recruiting it to the mem-
brane) and CD45 (which dephosphorylates Lck inhibi-
tory regulatory site) are required for the activation of the
essential kinase Lck.
Next we performed a logical steady state analysis for the
different time scales given above. These simulations pro-
vide a rough approximation to the dynamics of the sign-
aling cascade. Figure 10 shows the particular situation in
the early-event scenario (τ = 1) as displayed in CellNetAn-
alyzer. Figure 13 summarizes the logical steady state values
of important components obtained for the three different
time scales. The blue line shows the case for
TCR+CD4+CD45 stimulation, whereas the dashed red
line represents the case when only TCR+CD45 is stimu-
lated in the input layer. Similar analysis can be performed
using different scenarios, for example, in a cell where a
certain element has been knocked-out.
Conclusion
In this contribution we have presented a collection of
methods for the functional analysis of the structure of cel-
lular signaling and regulatory networks. As discussed in
the theoretical sections, different abstractions and formal-
isms can be used to encode and analyze the topology of
interaction networks. The simplest representations are
interaction graphs, which are restricted to one-to-one rela-
tionships but do yet capture important functional and
causal dependencies in the system under study. We have
shown that arguably the most important features of inter-
action graphs, namely feedback circuits and signaling (or
influence) pathways, can systematically be identified by
the concept and algorithm of elementary modes known
from stoichiometric (metabolic) network analysis. Feed-
back cycles are mainly responsible for the dynamic behav-
ior of the system, whereas signaling paths reveal network-
wide dependencies between species. In some cases, analy-
sis of feedback cycles and signaling paths may allow one
to predict unambiguously the qualitative effect upon per-
turbations of certain species (independently of kinetic
parameters and mechanisms). Falsification experiments
may then be used to identify missing or incorrect interac-
tions. Knowledge on all the signaling paths also facilitates
a systematic identification of optimal intervention strate-
gies. Again, a concept known from metabolic networks,
minimal cut sets, can be adapted and employed here.
However, inhibitory actions make this kind of analysis
more complicated and we therefore generalized the for-
Dependency matrix for the T-cell model for the early event
scenario (τ = 1: the feedback loops are not active)
Figure 12
Dependency matrix for the T-cell model for the early event
scenario (τ = 1: the feedback loops are not active). The meaning
of the different colors is the same as in Figure 6.
Dependency matrix for the T-cell model
Figure 11
Dependency matrix for the T-cell model. The meaning of the
different colors is the same as in Figure 6.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 24 of 26
(page number not for citation purposes)
malism of minimal cut sets leading to minimal interven-
tion sets.
The applicability of tools from metabolic network analy-
sis to interaction graphs relies on the fact that metabolic
networks are hypergraphs, which in turn are generaliza-
tions of graphs. In our opinion, the importance of hyper-
graphs in structural analyses of cellular interaction
networks has been underestimated. In fact, whenever
AND-connections occur in interactions of species, hyper-
graphical approaches become essential.
Boolean networks describe interaction networks in a more
constrained and deterministic manner than interaction
graphs, enabling discrete simulations. Herein we have
demonstrated that signed directed hypergraphs are capa-
ble to represent the logical structure of any Boolean net-
work. The hypergraphical coding of Boolean networks,
which relies on the sum-of-product representation of
Boolean networks (using only AND, OR and NOT opera-
tions), has several advantages: it is rather intuitive, it
mostly corresponds to the underlying molecular mecha-
nisms, and it is easy to store and to handle. A hypergraph-
ical representation of a Boolean network also establishes
a direct link to the corresponding (underlying) interaction
graph which can easily be derived from the hypergraph.
Finally, it facilitates a logical signal flow (or steady state)
analysis in Boolean networks which, as demonstrated in
this report, is useful for studying and predicting the qual-
itative input-output behavior of signaling networks with
respect to a given, possibly incomplete, set of initial state
values. This can be achieved here without an explicit enu-
meration and/or simulation of all possible trajectories.
In general, Boolean networks rely on stronger assump-
tions and knowledge than interaction graphs and a pure
logical description of all interactions is not always possi-
ble. We have suggested extensions of the Boolean frame-
work, such as incomplete truth tables of logical
operations, to handle these problems.
As pointed out by many authors (e.g. [67-69]) the logical
description and analysis of large signaling networks has a
strong relationship to electrical circuit analysis; however,
there still seems to be a large potential in employing the-
oretical and software tools from electrical engineering and
Boolean logic for investigating interaction networks. Sig-
nal flow analysis as introduced herein might be another
step in this direction.
Describing signal and mass flows equivalently as interac-
tions, as done herein, offers high flexibility and enables
one to integrate several types of cellular networks (such as
metabolic, signalling or regulatory ones) into one frame-
work. However, the higher level of abstraction comes with
the price that some molecular mechanisms are not always
precisely represented, as, for instance, the stoichiometric
coefficients in mass flows.
The potential of the introduced methods were demon-
strated on a model of a small part of the signaling machin-
ery of T-cells. The size and complexity of the model was
chosen so that the methods could be tested on a case study
of real size and complexity, while at the same time the
results could be (at least in part) intuitively understood
and proofed. If enough information is available, similar
models could be set up for any other signaling network.
Certainly, these tools will be especially useful in larger
interaction networks. Our current and future work aims to
expand and subsequently analyse the T-cell model, with
hopes that further understanding of this complex network
can improve current knowledge about important ill-
nesses, such as autoimmune diseases and leukemia. This
is certainly a challenging task, but the potential described
here makes it a worthy endeavour.
Availability and requirements
For academic purposes,CellNetAnalyzer can be obtained
for free via the website
http://www.mpi-magdeburg.mpg.de/projects/cna/
cna.html
Note that CellNetAnalyzer requires MATLAB® version 6.1
or higher.
Simulation results of LSS analysis of key elements of the T-
cell model using the two time-scales explained in the text
Figure 13
Simulation results of LSS analysis of key elements of the T-
cell model using the two time-scales explained in the text.
Blue line: upon TCR+CD4+CD45 activation; dashed red line:
only TCR+CD45 activation.
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 25 of 26
(page number not for citation purposes)
List of abbreviations
LIH: logical interaction hypergraph
LSS(s): logical steady state(s)
MCS(s): minimal cut set(s)
MIS(s): minimal intervention set(s)
Authors' contributions
SK elaborated the framework and the methods for study-
ing interaction graphs and logical interaction hypergraphs
and implemented algorithms in CellNetAnalyzer. JSR
mainly constructed the logical model of T-cell signalling
and he also contributed to the methods' development. JL
and LS assisted in the construction of the T-cell model.
EDG initiated the project on methods for structural anal-
ysis of signaling networks. SK and JSR prepared the man-
uscript jointly. All authors have read and accepted the
manuscript.
Acknowledgements
The work was supported by grants from the Deutsche Forschungsgemein-
schaft (FOR521) and Bundesministerium für Bildung und Forschung. Thanks
to the signaling group at the Institute of Immunology, especially to B.
Schraven, for helpful discussions on the T-cell signaling model and to J. Gag-
neur, J. Behre, U.U. Haus, R. Weismantel and Annegret Wagler for fruitful
discussions on theoretical issues. We thank R. Hemenway for critical read-
ing the manuscript.
References
1.
Downward J: The ins and outs of signalling. Nature 2001,
411:759-762.
2.
Papin JA, Hunter T, Palsson BO, Subramaniam S: Reconstruction of
cellular signalling networks and analysis of their properties.
Nat Rev Mol Cell Biol 2005, 6:99-111.
3.
Kholodenko BN: Negative feedback and ultrasensitivity can
bring about oscillations in the mitogen-activated protein
kinase cascades. Eur J Biochem 2000, 267:1583-8.
4.
Schoeberl B, Eichler-Jonsson C, Gilles ED, Müller G: Computa-
tional modeling of the dynamics of the MAP kinase cascade
activated by surface and internalized EGF receptors. Nat Bio-
tech 2002, 20:370-375.
5.
Sauro HM, Kholodenko BN: Quantitative analysis of signaling
networks. Biophysics & Molecular Biology 2004, 86:5-43.
6.
Sasagawa S, Ozaki Y, Fujita K, Kuroda S: Prediction and validation
of the distinct dynamics of transient and sustained ERK acti-
vation. Nat Cell Biol 2005, 7:365-373.
7.
Styczynski MP, Stephanopoulos G: Overview of computational
methods for the inference of gene regulatory networks. Com-
puters & Chemical Engineering 2005, 29:519-534.
8.
Sachs K, Perez O, Pe'er D, Lauffenburger D, Nolan GP: Causal pro-
tein signaling networks dreived from multiparamter single-
cell data. Science 2005, 308:523-529.
9.
de la Fuente A, Brazhnik P, Mendes P: Linking the genes: inferring
quantitative gene networks from microarray data. Trends
Genet 2002, 18:395-398.
10.
Sontag E, Kiyatkin A, Kholodenko BN: Inferring dynamic archi-
tecture of cellular networks using time series of gene expres-
sion, protein and metabolite data. Bioinformatics 2004,
20:1877-1886.
11.
Price ND, Reed JL, Palsson BO: Genome-scale models of micro-
bial cells: evaluating the consequences of constraints. Nat Rev
Microbiol 2004, 2:886-897.
12.
Oda K, Matsuoka Y, Funahashi A, Kitano H: A comprehensive
pathway map of epidermal growth factor receptor signaling.
Molecular Systems Biology . 10.1038/msb4100014
13.
Jeong H, Mason SP, Barabasi AL, Oltvai ZN: Lethality and central-
ity in protein networks. Nature 2001, 411:41-42.
14.
Papin JA, Palsson BO: Topological analysis of mass-balanced sig-
naling networks: a framework to obtain emergent network
properties including crosstalk. J Theor Biol 2004, 227:283-297.
15.
Papin JA, Palsson BO: The JAK-STAT Signaling Network in the
Human B-Cell: An Extreme Signaling Pathway Analysis. Bio-
phys J 2004, 87:37-46.
16.
Zevedei-Oancea I, Schuster S: A theoretical framework for
detecting signal transfer routes in signalling networks. Com-
puters & Chemical Engineering 2005, 29:597-617.
17.
Binder B, Heinrich R: Interrelations between dynamical prop-
erties and structural characteristics of signal transduction
networks. Genome Inform 2004, 15:13-23.
18.
Wagner A, Wright J: Compactness and cycles in signal trans-
duction and transcriptional regulation networks: a signature
of natural selection? Advances in Complex Systems 2005, 7:419-432.
19.
Kaufman M, Andris F, Leo O: A logical analysis of T cell activa-
tion and anergy. Proc Natl Acad Sci U S A 1999, 96:3894-3899.
20.
Mendoza L, Thieffry D, Alvarez-Buylla ER: Genetic control of
flower morphogenesis in Arabidopsis thaliana: a logical anal-
ysis. Bioinformatics 1999, 15:593-606.
21.
Thomas R, D'Ari R: Biological feedback Boca Raton: CRC Press; 1990.
22.
Albert R, Othmer HG: The topology of the regulatory interac-
tions predicts the expression pattern of the Drosophila seg-
ment polarity genes. J Theor Biology 2003, 223:1-18.
23.
Espinosa-Soto C, Padilla-Longoria P, Alvarez-Buylla ER: A gene reg-
ulatory network model for cell-fate determination during
Arabidopsis thaliana flower development that is robust and
recovers experimental gene expression profiles. Plant Cell
2004, 16:2923-2939.
24.
Gagneur J, Casari G: From molecular networksto qualitative
cell behavior. FEBS Letter 2005, 579:1867-1871.
25.
Schuster S, Fell DA, Dandekar T: A general definition of meta-
bolic pathways useful for systematic organization and analy-
sis of complex metabolic networks. Nat Biotechnol 2000,
18:326-332.
26.
Stelling J, Klamt S, Bettenbrock K, Schuster S, Gilles ED: Metabolic
network structure determines key aspects of functionality
and regulation. Nature 2002, 420:190-193.
27.
Kauffman KJ, Prakash P, Edwards JS: Advances in flux balance
analysis. Curr Opin Biotechnol 2003, 14:491-496.
28.
Klamt S, Gilles ED: Minimal cut sets in biochemical reaction
networks. Bioinformatics 2004, 20:226-234.
29.
Klamt S: Generalised concept of minimal cut sets in biochem-
ical networks. Biosystems 2006, 83:233-247.
30.
Schlessinger J: Cell signaling by receptor tyrosine kinases. Cell
2000, 103:211-25.
31.
de Jong H: Modeling and simulation of genetic regulatory sys-
tems: a literature overview. J Comp Biol 2002, 9:67-103.
32.
Gross JL, Yellen J: Handbook of graph theory Boca Raton: CRC Press;
2004.
33.
Heinrich R, Schuster S: The Regulation of Cellular Systems New York:
Chapman & Hall; 1996.
34.
Bollabas B: Modern graph theory New York, Springer-Verlag; 1998.
35.
Thomas R, Kaufman M: Multistationarity, the basis of cell differ-
entiation and memory. I Structural conditions of multista-
tionarity and other non-trivial behavior. Chaos 2001,
11:170-179.
36.
Angeli D, Ferrell JE, Sontag ED: Detection of multistability, bifur-
cations and hysteresis in a large class of biological positive-
feedback systems. PNAS 2004, 101:1822-1827.
37.
Reth M, Brummer T: Feedback regulation of lymphocyte signal-
ling. Nat Rev Immunol 2004, 4:269-77.
38.
Xiong W, Ferrel JR Jr: A positive-feedback-based bistable
'memory module' that governs a cell fate decision. Nature
2003, 426:460-5.
39.
Soule C: Graphic requirements for multistationarity. Com-
PlexUs 2003, 1:123-133.
40.
Tarjan R: Enumeration of the elementary circuits of a
directed graph. SIAM J Comput 1973, 2:211-216.
41.
Gleiss PM, Stadler PF, Wagner A, Fell DA: Relevant cycles in
chemical reaction networks. Adv Complex S 2001, 4:207-226.
Publish with BioMed Central and every
scientist can read your work free of charge
"BioMed Central will be the most significant development for
disseminating the results of biomedical research in our lifetime."
Sir Paul Nurse, Cancer Research UK
Your research papers will be:
available free of charge to the entire biomedical community
peer reviewed and published immediately upon acceptance
cited in PubMed and archived on PubMed Central
yours — you keep the copyright
Submit your manuscript here:
http://www.biomedcentral.com/info/publishing_adv.asp
BioMedcentral
BMC Bioinformatics 2006, 7:56
http://www.biomedcentral.com/1471-2105/7/56
Page 26 of 26
(page number not for citation purposes)
42.
Lawler E: Combinatorial Optimization – Networks and Matroids Mineola:
Dover Publications; 2001.
43.
Klamt S, Gagneur J, Kamp A: Algorithmic approaches for com-
puting elementary modes in large biochemical reaction net-
works. IEE Proceedings Systems Biology 2005, 152(4):249-255.
44.
Gagneur J, Klamt S: Computation of elementary modes: a uni-
fying framework and the new binary approach. BMC Bioinfor-
matics 2004, 5:175.
45.
Xiong M, Jinying Z, Xiong H: Network-based regulatory path-
ways analysis. Bioinformatics 2004, 20:2056-2066.
46.
Urbanczik R, Wagner C: An improved algorithm for stoichio-
metric network analysis: theory and applications. Bioinformat-
ics 2005, 21:1203-1210.
47.
Pfeiffer T, Sánchez-Valdenebro I, Nuño JC, Montero F, Schuster S:
METATOOL: For studying metabolic networks. Bioinformatics
1999, 15:251-257.
48.
Newman MEJ: Scientific collaboration networks: II. Shortest
paths, weighted networks and centrality. Phys Rev E 2001,
64:016132.
49.
Zeigarnik AV: On hypercycles and hypercircuits in hyper-
graphs. In Discrete Mathematical Chemistry Volume 51. Edited by:
Hansen P, Fowler PW, Zheng M. DIMACS series in discrete mathe-
matics and theoretical computer science; 2000:377-383.
50.
Mendelson E: Schaum's outline of Boolean algebra and switching circuits
McGraw-Hill, New York; 1970.
51.
Kauffman SA: Metabolic stability and epigenesis in randomly
constructed genetic nets. J Theor Biol 1969, 22:437-467.
52.
Devloo V, Hansen P, Labbe M: Identification of all steady states
in large networks by logical analysis. Bulletin of Mathematical Biol-
ogy 2003, 65:1025-1051.
53.
Stephanopoulos GN, Aristidou AA, Nielsen J: Metabolic Engineering
Academic Press, San Diego; 1998.
54.
Klamt S, Schuster S, Gilles ED: Calculability analysis in underde-
termined metabolic networks illustrated by a model of the
central metabolism in purple nonsulfur bacteria. Biotechnol
Bioeng 2002, 77:734-751.
55.
Ebenhöh O, Handorf T, Heinrich R: Structural analysis of
expanding metabolic networks. Genome Informatics 2004,
15:35-45.
56.
Klamt S, Stelling J, Ginkel M, Gilles ED: FluxAnalyzer: exploring
structure, pathways, and flux distributions in metabolic net-
works on interactive flux maps. Bioinformatics 2003, 19:261-269.
57.
[http://www.mpi-magdeburg.mpg.de/projects/cna/cna.html].
58.
Benjamini E, Coico R, Sunshine G: Immunology-A short course Wiley-
Liss; 2000.
59.
Huang Y, Wange RL: T cell receptor signaling: beyond complex
complexes. J Biol Chem 2004, 279:28827-30.
60.
Togni M, Lindquist J, Gerber A, Kolsch U, Hamm-Baarke A, Kliche S,
Schraven B: The role of adaptor proteins in lymphocyte acti-
vation. Mol Immunol 2004, 41:615-630.
61.
Horejsi V, Zhang W, Schraven B: Transmembrane adaptor pro-
teins: organizers of immunoreceptor signalling. Nat Rev Immu-
nol 2004, 4:603-16.
62.
Torgersen KM, Vang T, Abrahamsen H, Yaqub S, Horejsi V, Schraven
B, Rolstad B, Mustelin T, Tasken K: Release from tonic inhibition
of T cell activation through transient displacement of C-ter-
minal Src kinase (Csk) from lipid rafts. J Biol Chem 2001,
276:29313-29318.
63.
Filipp D, Julius M: Lipid rafts: resolution of the "fyn problem"?
Mol Immunol 2004, 41:645-56.
64.
Schaeffer EM, Debnath J, Yap G, McVicar D, Liao XC, Littman DR,
Sher A, Varmus HE, Lenardo MJ, Schwartzberg PL: Requirement
for Tec kinases Rlk and Itk in T cell receptor signaling and
immunity. Science 1999, 284:638-41.
65.
Roose JP, Mollenauer M, Gupta VA, Stone J, Weiss A: A diacylglyc-
erol-protein kinase C-RasGRP1 pathway directs Ras activa-
tion upon antigen receptor stimulation of T cells. Mol Cell Biol
2005, 25:4426-41.
66.
Duan L, Reddi AL, Ghosh A, Dimri M, Band H: The Cbl Family and
Other Ubiquitin Ligases Destructive Forces in Control of
Antigen Receptor Signaling. Immunity 2004, 21:7-17.
67.
Genoud T, Santa Cruz MBT, Metraux JP: Numeric simulation of
plant signaling networks. Plant Physiology 2001, 126:1430-1437.
68.
Hasty J, McMillen D, Collins JJ: Engineered gene circuits. Nature
2002, 420:224-230.
69.
Lok L: Software for signaling networks, electronic and cellu-
lar. Science's STKE 2002, 122:PE11.
|
16464248
|
Lck = ( ( CD45 AND ( ( ( CD8 ) ) ) ) AND NOT ( PAGCsk ) )
ZAP70 = ( ( TCRphos AND ( ( ( Lck ) ) ) ) AND NOT ( cCbl ) )
Itk = ( ZAP70 AND ( ( ( SLP76 ) ) ) )
SEK = ( PKCth )
RasGRP1 = ( PKCth AND ( ( ( DAG ) ) ) )
SLP76 = ( Gads )
CREB = ( Rsk )
Rlk = ( Lck )
ERK = ( MEK )
Gads = ( LAT )
IkB = NOT ( ( IKKbeta ) )
Ca = ( IP3 )
Ras = ( RasGRP1 ) OR ( Grb2Sos )
IP3 = ( PLCg_act )
PAGCsk = ( Fyn )
JNK = ( SEK )
Jun = ( JNK )
Fos = ( ERK )
TCRphos = ( Fyn )
TCRbind = ( ( TCRlig ) AND NOT ( cCbl ) )
NFAT = ( Calcin )
Raf = ( Ras )
PKCth = ( DAG )
Grb2Sos = ( LAT )
AP1 = ( Fos AND ( ( ( Jun ) ) ) )
PLCg_act = ( ZAP70 AND ( ( ( SLP76 AND PLCg_bind AND Rlk ) ) ) ) OR ( Itk AND ( ( ( SLP76 AND ZAP70 AND PLCg_bind ) ) ) )
cCbl = ( ZAP70 )
IKKbeta = ( PKCth )
LAT = ( ZAP70 )
Nfkb = NOT ( ( IkB ) )
PLCg_bind = ( LAT )
Fyn = ( TCRbind AND ( ( ( CD45 ) ) ) ) OR ( CD45 AND ( ( ( Lck ) ) ) )
Calcin = ( Ca )
DAG = ( PLCg_act )
MEK = ( Raf )
Rsk = ( ERK )
CRE = ( CREB )
|
BioMed Central
Page 1 of 18
(page number not for citation purposes)
Theoretical Biology and Medical
Modelling
Open Access
Research
A method for the generation of standardized qualitative dynamical
systems of regulatory networks
Luis Mendoza* and Ioannis Xenarios
Address: Serono Pharmaceutical Research Institute, 14, Chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
Email: Luis Mendoza* - luis.mendoza@serono.com; Ioannis Xenarios - ioannis.xenarios@serono.com
* Corresponding author
Abstract
Background: Modeling of molecular networks is necessary to understand their dynamical
properties. While a wealth of information on molecular connectivity is available, there are still
relatively few data regarding the precise stoichiometry and kinetics of the biochemical reactions
underlying most molecular networks. This imbalance has limited the development of dynamical
models of biological networks to a small number of well-characterized systems. To overcome this
problem, we wanted to develop a methodology that would systematically create dynamical models
of regulatory networks where the flow of information is known but the biochemical reactions are
not. There are already diverse methodologies for modeling regulatory networks, but we aimed to
create a method that could be completely standardized, i.e. independent of the network under
study, so as to use it systematically.
Results: We developed a set of equations that can be used to translate the graph of any regulatory
network into a continuous dynamical system. Furthermore, it is also possible to locate its stable
steady states. The method is based on the construction of two dynamical systems for a given
network, one discrete and one continuous. The stable steady states of the discrete system can be
found analytically, so they are used to locate the stable steady states of the continuous system
numerically. To provide an example of the applicability of the method, we used it to model the
regulatory network controlling T helper cell differentiation.
Conclusion: The proposed equations have a form that permit any regulatory network to be
translated into a continuous dynamical system, and also find its steady stable states. We showed
that by applying the method to the T helper regulatory network it is possible to find its known
states of activation, which correspond the molecular profiles observed in the precursor and
effector cell types.
Background
The increasing use of high throughput technologies in dif-
ferent areas of biology has generated vast amounts of
molecular data. This has, in turn, fueled the drive to incor-
porate such data into pathways and networks of interac-
tions, so as to provide a context within which molecules
operate. As a result, a wealth of connectivity information
is available for multiple biological systems, and this has
been used to understand some global properties of bio-
logical networks, including connectivity distribution [1],
recurring motifs [2] and modularity [3]. Such informa-
tion, while valuable, provides only a static snapshot of a
Published: 16 March 2006
Theoretical Biology and Medical Modelling2006, 3:13
doi:10.1186/1742-4682-3-13
Received: 12 December 2005
Accepted: 16 March 2006
This article is available from: http://www.tbiomed.com/content/3/1/13
© 2006Mendoza and Xenarios; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 2 of 18
(page number not for citation purposes)
network. For a better understanding of the functionality of
a given network it is important to study its dynamical prop-
erties. The consideration of dynamics allows us to answer
questions related to the number, nature and stability of
the possible patterns of activation, the contribution of
individual molecules or interactions to establishing such
patterns, and the possibility of simulating the effects of
loss- or gain-of-function mutations, for example.
Mathematical modeling of metabolic networks requires
specification of the biochemical reactions involved. Each
reaction has to incorporate the appropriate stoichiometric
coefficients to account for the principle of mass conserva-
tion. This characteristic simplifies modeling, because it
implies that at equilibrium every node of the metabolic
network has a total mass flux of zero [4,5]. There are cases,
however, where the underlying biochemical reactions are
not known for large parts of a pathway, but the direction
of the flow of information is known, which is the case for
so-called regulatory networks (see for example [6,7]). In
these cases, the directionality of signaling is sufficient for
developing mathematical models of how the patterns of
activation and inhibition determine the state of activation
of the network (for a review, see [8]).
When cells receive external stimuli such as hormones,
mechanical forces, changes in osmolarity, membrane
potential etc., there is an internal response in the form of
multiple intracellular signals that may be buffered or may
eventually be integrated to trigger a global cellular
response, such as growth, cell division, differentiation,
apoptosis, secretion etc. Modeling the underlying molec-
ular networks as dynamical systems can capture this chan-
neling of signals into coherent and clearly identifiable
Methodology
Figure 1
Methodology. Schematic representation of the method for systematically constructing a dynamical model of a regulatory net-
work and finding its stable steady states.
(t))
(t)...x
g(x
)
(t
x
n
i
1
1
Convert the network
into a discrete dynamical
system
Find all the stable steady
states with the generalized
logical analysis
)
...x
f(x
dt
dx
n
i
1
Convert the network
into a continuous
dynamical system
...
1
)
(
;0
)
(
0
2
0
1
t
x
t
x
Use the steady states of the
discrete system as initial
states to solve numerically
the continuous system
Let the continuous system run
until it converges to a steady state
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 3 of 18
(page number not for citation purposes)
stable cellular behaviors, or cellular states. Indeed, quali-
tative and semi-quantitative dynamical models provide
valuable information about the global properties of regu-
latory networks. The stable steady states of a dynamical
system can be interpreted as the set of all possible stable
patterns of expression that can be attained within the par-
ticular biological network that is being modeled. The
advantages of focusing the modeling on the stable steady
states of the network are two-fold. First, it reduces the
quantity of experimental data required to construct a
model, e.g. kinetic and rate limiting step constants,
because there is no need to describe the transitory
response of the network under different conditions, only
the final states. Second, it is easier to verify the predictions
of the model experimentally, since it requires stable cellu-
lar states to be identified; that is, long-term patterns of
activation and not short-lived transitory states of activa-
tion that may be difficult to determine experimentally.
In this paper we propose a method for generating qualita-
tive models of regulatory networks in the form of contin-
uous dynamical systems. The method also permits the
stable steady states of the system to be localized. The pro-
cedure is based on the parallel construction of two
dynamical systems, one discrete and one continuous, for
the same network, as summarized in Figure 1. The charac-
teristic that distinguishes our method from others used to
model regulatory networks (as summarized in [8]) is that
the equations used here, and the method deployed to ana-
lyze them, are completely standardized, i.e. they are not
network-specific. This feature permits systematic applica-
tion and complete automation of the whole process, thus
The Th network
Figure 2
The Th network. The regulatory network that controls the differentiation process of T helper cells. Positive regulatory
interactions are in green and negative interactions in red.
IFN-γ
IL-4
SOCS1
IL-12R
IFN-γR
IL-4R
JAK1
STAT4
STAT6
GATA3
T-bet
IL-12
IL-18
IL-18R
IRAK
IFN-βR
IFN-β
IL-10
IL-10R
STAT3
STAT1
NFAT
TCR
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 4 of 18
(page number not for citation purposes)
speeding up the analysis of the dynamical properties of
regulatory networks. Moreover, in contrast to methodolo-
gies for the automatic analysis of biochemical networks
(as in [9]; for example), our method can be applied to net-
works for which there is a lack of stoichiometric informa-
tion. Indeed, the method requires as sole input the
information regarding the nature and directionality of the
regulatory interactions. We provide an example of the
applicability of our method, using it to create a dynamical
model for the regulatory network that controls the differ-
entiation of T helper (Th) cells.
Results and discussion
Equations 1 and 3 (see Methods) provide the means for
transforming a static graph representation of a regulatory
network into two versions of a dynamical system, a dis-
crete and a continuous description, respectively. As an
example, we applied these equations to the Th regulatory
network, shown in Figure 2. Briefly, the vertebrate
immune system contains diverse cell populations, includ-
ing antigen presenting cells, natural killer cells, and B and
T lymphocytes. T lymphocytes are classified as either T
helper cells (Th) or T cytotoxic cells (Tc). T helper cells
take part in cell- and antibody-mediated immune
responses by secreting various cytokines, and they are fur-
ther sub-divided into precursor Th0 cells and effector Th1
and Th2 cells, depending on the array of cytokines that
they secrete [10]. The network that controls the differenti-
ation from Th0 towards the Th1 or Th2 phenotypes is
rather complex, and discrete modeling has been used to
understand its dynamical properties [11,12]. In this work
we used an updated version of the Th network, the molec-
ular basis of which is included in the Methods. Also, we
implement for the first time a continuous model of the Th
network.
By applying Equation 1 to the network in Figure 2, we
obtained Equation 2, which constitutes the discrete ver-
sion of the dynamical system representing the Th net-
work. Similarly, the continuous version of the Th network
was obtained by applying Equation 3 to the network in
Figure 2. In this case, however, some of the resulting equa-
tions are too large to be presented inside the main text, so
we included them as the Additional file 1. Moreover,
instead of just typing the equations, we decided to present
them in a format that might be used directly to run simu-
lations. The continuous dynamical system of the Th net-
work is included as a plain text file that is able to run on
the numerical computation software package GNU
Octave http://www.octave.org.
The high non-linearity of Equation 3 implies that the con-
tinuous version of the dynamical model has to be studied
numerically. In contrast, the discrete version can be stud-
Table 1: Stable steady states of the dynamical systems. a
DISCRETE SYSTEM
CONTINUOUS SYSTEM
Th0
Th1
Th2
Th0
Th1
Th2
GATA3
0
0
1
0
0
1
IFN-β
0
0
0
0
0
0
IFN-βR
0
0
0
0
0
0
IFN-γ
0
1
0
0
0.71443
0
IFN-γR
0
1
0
0
0.9719
0
IL-10
0
0
1
0
0
1
IL-10R
0
0
1
0
0
1
IL-12
0
0
0
0
0
0
IL-12R
0
0
0
0
0
0
IL-18
0
0
0
0
0
0
IL-18R
0
0
0
0
0
0
IL-4
0
0
1
0
0
1
IL-4R
0
0
1
0
0
1
IRAK
0
0
0
0
0
0
JAK1
0
0
0
0
0.00489
0
NFAT
0
0
0
0
0
0
SOCS1
0
1
0
0
0.89479
0
STAT1
0
0
0
0
0.00051
0
STAT3
0
0
1
0
0
1
STAT4
0
0
0
0
0
0
STAT6
0
0
1
0
0
1
T-bet
0
1
0
0
0.89479
0
TCR
0
0
0
0
0
0
a. Homologous non-zero values between the discrete and the continuous systems are shown in bold
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 5 of 18
(page number not for citation purposes)
ied analytically by using generalized logical analysis,
allowing all its stable steady states to be located (see Meth-
ods). In our example, the discrete system described by
Equation 2 has three stable steady states (see Table 1).
Importantly, these states correspond to the molecular pro-
files observed in Th0, Th1 and Th2 cells. Indeed, the first
stable steady state reflects the pattern of Th0 cells, which
are precursor cells that do not produce any of the
cytokines included in the model (IFN-β, IFN-γ, IL-10, IL-
12, IL-18 and IL-4). The second steady state represents
Th1 cells, which show high levels of activation for IFN-γ,
IFN-γR, SOCS1 and T-bet, and with low (although not
zero) levels of JAK1 and STAT1. Finally, the third steady
state corresponds to the activation observed in Th2 cells,
with high levels of activation for GATA3, IL-10, IL-10R, IL-
4, IL-4R, STAT3 and STAT6.
Equation 3 defines a highly non-linear continuous
dynamical system. In contrast with the discrete system,
these continuous equations have to be studied numeri-
cally. Numerical methods for solving differential equa-
tions require the specification of an initial state, since they
proceed via iterations. In our method, we propose to use
the stable steady states of the discrete system as the initial
states to solve the continuous system that results from
application of equation 3 to a given network. We used a
standard numerical simulation method to solve the con-
tinuous version of the Th model (see Methods). Starting
alternatively from each of the three stable steady states
found in the discrete model, i.e. the Th0, Th1 and Th2
states, the continuous system was solved numerically
until it converged. The continuous system converged to
values that could be compared directly with the stable
steady states of the discrete system (Table 1). Note that the
Th0 and Th2 stable steady states fall in exactly the same
position for both the discrete and the continuous dynam-
ical systems, and in close proximity for the Th1 state. This
finding highlights the similarity in qualitative behavior of
the two models constructed using equations 1 and 3,
despite their different mathematical frameworks.
Despite the qualitative similarity between the discrete and
continuous systems, there is no guarantee that the contin-
uous dynamical system has only three stable steady states;
there might be others without a counterpart in the discrete
system. To address this possibility, we carried out a statis-
tical study by finding the stable steady states reached by
the continuous system starting from a large number of ini-
Table 2: Regions of the state space reached by the continuous version of the Th model, as revealed by a large number of simulations
starting from a random initial state. a
Th0
Th1
Th2
Avrg.
Std. Dev.
Avrg.
Std. Dev.
Avrg.
Std. Dev.
GATA3
0.00003
0.00008
0.00000
0.00000
0.99997
0.00007
IFN-β
0.00000
0.00000
0.00000
0.00000
0.00000
0.00000
IFN-βR
0.00000
0.00001
0.00000
0.00001
0.00000
0.00001
IFN-γ
0.00005
0.00013
0.71438
0.00059
0.00000
0.00001
IFN-γR
0.00004
0.00011
0.97169
0.00040
0.00001
0.00004
IL-10
0.00003
0.00007
0.00000
0.00001
0.99999
0.00004
IL-10R
0.00005
0.00010
0.00000
0.00001
0.99999
0.00002
IL-12
0.00000
0.00001
0.00000
0.00000
0.00000
0.00001
IL-12R
0.00000
0.00002
0.00000
0.00001
0.00000
0.00001
IL-18
0.00000
0.00001
0.00000
0.00000
0.00000
0.00001
IL-18R
0.00000
0.00002
0.00000
0.00001
0.00000
0.00001
IL-4
0.00002
0.00006
0.00000
0.00001
0.99995
0.00011
IL-4R
0.00002
0.00004
0.00000
0.00001
0.99990
0.00022
IRAK
0.00001
0.00005
0.00000
0.00003
0.00001
0.00004
JAK1
0.00002
0.00008
0.00487
0.00005
0.00001
0.00005
NFAT
0.00001
0.00003
0.00000
0.00002
0.00001
0.00003
SOCS1
0.00009
0.00022
0.89486
0.00037
0.00002
0.00006
STAT1
0.00001
0.00005
0.00051
0.00003
0.00002
0.00005
STAT3
0.00012
0.00023
0.00001
0.00002
1.00000
0.00002
STAT4
0.00001
0.00003
0.00000
0.00003
0.00000
0.00001
STAT6
0.00001
0.00004
0.00000
0.00002
0.99990
0.00023
T-bet
0.00007
0.00018
0.89485
0.00036
0.00000
0.00000
TCR
0.00000
0.00001
0.00000
0.00000
0.00000
0.00001
a. Only three regions of the activation space were found in the continuous Th model after running it from 50,000 different random initial states. The
average and standard deviations of all the results are shown. All variables had a random initial state in the closed interval [0,1]. From the 50,000
simulations, 8195 (16.39%) converged to the Th0 state, 25575 (51.15%) to the Th1 state, and 16230 (32.46%) to the Th2 state. Bold numbers as in
Table 1.
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 6 of 18
(page number not for citation purposes)
Stability of the steady states of the continuous model of the Th network
Figure 3
Stability of the steady states of the continuous model of the Th network. a. The Th0 state is stable under small per-
turbations. b. A large perturbation on IFN-γ is able to move the system from the Th0 to the Th1 steady state. This latter state
is stable to perturbations. c. A large perturbation of IL-4 moves the system from the Th0 state to the Th2 state, which is sta-
ble. For clarity, only the responses of key cytokines and transcription factors are plotted. The time is represented in arbitrary
units.
level of activation
level of activation
level of activation
a
c
b
IFN-γ perturbation
IL-4 perturbation
IFN-γ perturbation
IFN-γ perturbation
IL-4 perturbation
IL-4 perturbation
time
time
time
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 7 of 18
(page number not for citation purposes)
tial states. The continuous system was run 50,000 times,
each time with the nodes in a random initial state within
the closed interval between 0 and 1. In all cases, the sys-
tem converged to one of only three different regions
(Table 2), corresponding to the above-mentioned Th0,
Th1 and Th2 states. These results still do not eliminate the
possibility that other stable steady states exist in the con-
tinuous system. Nevertheless, they show that if such addi-
tional stable steady states exist, their basin of attractions is
relatively small or restricted to a small region of the state
space.
The three steady states of the continuous system are stable,
since they can resist small perturbations, which create
transitory responses that eventually disappear. Figure 3a
shows a simulation where the system starts in its Th0 state
and is then perturbed by sudden changes in the values of
IFN-γ and IL-4 consecutively. Note that the system is capa-
ble of absorbing the perturbations, returning to the origi-
nal Th0 state. If a perturbation is large enough, however,
it may move the system from one stable steady state to
another. If the system is in the Th0 state and IFN-γ is tran-
siently changed to it highest possible value, namely 1, the
whole system reacts and moves to its Th1 state (Figure
3b). A large second perturbation by IL-4, now occurring
when the system is in its Th1 state, does not push the sys-
tem into another stable steady state, showing the stability
of the Th1 state. Conversely, if the large perturbation of IL-
4 occurs when the system is in the Th0 state, it moves the
system towards the Th2 state (Figure 3c). In this case, a
second perturbation, now in IFN-γ, creates a transitory
response that is not strong enough to move the system
away from the Th2 state, showing the stability of this
steady state. These changes from one stable steady state to
another reflect the biological capacities of IFN-γ and IL-4
to act as key signals driving differentiation from Th0
towards Th1 and Th2 cells, respectively[10]. Furthermore,
note that the Th1 and Th2 steady states are more resistant
to large perturbations than the Th0 state, a characteristic
that represents the stability of Th1 and Th2 cells under dif-
ferent experimental conditions.
Alternative Th network
Figure 6
Alternative Th network. T helper pathway published in
[43], reinterpreted as a signaling network.
IL-12
IL-4
STAT1
IL-12R
STAT4
T-bet
IFN-γ
IFN-γR
IL-4R
STAT6
GATA3
IL-5
IL-13
TCR
Alternative Th network
Figure 4
Alternative Th network. T helper pathway published in
[69], reinterpreted as a signaling network.
IL-12
Steroids
IFN-γ
Inf.
Resp.
IL-4
IL-5
IL-10
Alternative Th network
Figure 5
Alternative Th network. T helper pathway published in
[70], reinterpreted as a signaling network.
IFN-γ
CSIF
IL-2
IL-4
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 8 of 18
(page number not for citation purposes)
The whole process resulted in the creation of a model with
qualitative characteristics fully comparable to those found
in the experimental Th system. Notably, the model used
default values for all parameters. Indeed, the continuous
dynamical system of the Th network has a total of 58
parameters, all of which were set to the default value of 1,
and one parameter (the gain of the sigmoids) with a
default value of 10. This set of default values sufficed to
capture the correct qualitative behavior of the biological
system, namely, the existence of three stable steady states
that represent Th0, Th1 and Th2 cells. Readers can run
simulations on the model by using the equations pro-
vided in the "Th_continuous_model.octave.txt" file. The
file was written to allow easy modification of the initial
states for the simulations, as well as the values of all
parameters.
Analysis of previously published regulatory networks
related to Th cell differentiation
We wanted to compare the results from our method (Fig-
ure 1) as applied to our proposed network (Figure 2) with
some other similar networks. The objective of this com-
parison is to show that our method imposes no restric-
tions on the number of steady states in the models.
Therefore, if the procedure is applied to wrongly recon-
structed networks, the results will not reflect the general
characteristics of the biological system. While there have
been multiple attempts to reconstruct the signaling path-
ways behind the process of Th cell differentiation, they
have all been carried out to describe the molecular com-
ponents of the process, but not to study the dynamical
behavior of the network. As a result, most of the schematic
representations of these pathways are not presented as
regulatory networks, but as collections of molecules with
different degrees of ambiguity to describe their regulatory
interactions. To circumvent this problem, we chose four
pathways with low numbers of regulatory ambiguities
and translated them as signaling networks (Figures 4
through 7).
The methodology introduced in this paper was applied to
the four reinterpreted networks for Th cell differentiation.
Alternative Th network
Figure 7
Alternative Th network. T helper pathway published in [71], reinterpreted as a signaling network.
Itk
NFAT
IL-18R
c-Maf
IL-4R
IL-13
STAT6
JNK2
IL-4
IL-5
IL-18
Lck
CD4
JNK
IRAK
NFkB
TRAF6
IFN-γ
T-bet
STAT4
GATA3
TCR
Ag/
MHC
IL-12R
IL-12
ATF2
p38/
MAPK
MKK3
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 9 of 18
(page number not for citation purposes)
The stable steady states of the resulting discrete and con-
tinuous models are presented in Tables 3 through 6.
Notice that none of these four alternative networks could
generate the three stable steady states representing Th0,
Th1 and Th2 cells. Two networks reached only two stable
steady states, while two others reached more than three.
Notably, all these four networks included one state repre-
senting the Th0 state, and at least one representing the
Th2 state. The absence of a Th1 state in two of the net-
works might reflect the lack of a full characterization of
the IFN-γ signaling pathway at the time of writing the cor-
responding papers.
It is important to note that the failure of these four alter-
native networks to capture the three states representing Th
cells is not attributable to the use of very simplistic and/or
outdated data. Indeed, the network in Figure 6 comes
from a relatively recent review, while that in Figure 7 is
rather complex and contains five more nodes than our
own proposed network (Figure 2). All this stresses the
importance of using a correctly reconstructed network to
develop dynamical models, either with our approach or
any other.
Conclusion
There is a great deal of interest in the reconstruction and
analysis of regulatory networks. Unfortunately, kinetic
information about the elements that constitute a network
or pathway is not easily gathered, and hence the analysis
of its dynamical properties (via simulation packages such
as [13]) is severely restricted to a small set of well-charac-
terized systems. Moreover, the translation from a static to
a dynamical representation normally requires the use of a
network-specific set of equations to represent the expres-
sion or concentration of every molecule in the system.
We herein propose a method for generating a system of
ordinary differential equations to construct a model of a
regulatory network. Since the equations can be unambig-
uously applied to any signaling or regulatory network, the
construction and analysis of the model can be carried out
systematically. Moreover, the process of finding the stable
steady states is based on the application of an analytical
method (generalized logical analysis [14,15] on a discrete
version of the model), followed by a numerical method
(on the continuous version) starting from specific initial
states (the results obtained from the logical analysis). This
characteristic allows a fully automated implementation of
our methodology for modeling. In order to construct the
equations of the continuous dynamical system with the
exclusive use of the topological information from the net-
work, the equations have to incorporate a set of default
values for all the parameters. Therefore, the resulting
model is not optimized in any sense. However, the advan-
tage of using Equation 3 is that the user can later modify
the parameters so as to refine the performance of the
Table 4: Stable steady states of the signaling network in Figure 5
Discrete state 1
Discrete state 2
Discrete state 3
Discrete state 4
Discrete state 5
Discrete state 6
Discrete state 7
CSIF
0
0
1
0
0.5
0.5
0
IFN-γ
0
1
0
0.5
0
0
0. 5
IL-2
0
1
0
0.5
0.5
0.5
0
IL-4
0
0
1
0.5
0
0.5
0.5
Continuous
state 1
Continuous
state 2
Continuous
state 3
Continuous
state 4
Continuous
state 5
Continuous
state 6
Continuous
state 7
CSIF
0
0.0034416
0.8888881
0.0034999
4.9132E-5
0.8881746
4.3001E-5
IFN-γ
0
0.8888881
0.0034416
0.8881746
4.300E-5
0.0034999
4.9132E-5
IL-2
0
0.8888881
0.0034416
0.8881746
4.3154E-5
0.0035227
4.8979E-5
IL-4
0
0.0034416
0.8888881
0.0035227
4.8979E-5
0.8881746
4.3154E-5
Table 3: Stable steady states of the signaling network in Figure 4
Discrete state 1
Discrete state 2
Continuous state 1
Continuous state 2
IFN-γ
0
0
0
0
IL-10
0
1
0
0.78995
IL-12
0
0
0
0
IL-4
0
1
0
0.89469
IL-5
0
0
0
0.01343
Inf. Resp.
0
0
0
0.00737
Steroids
0
0
0
0.00105
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 10 of 18
(page number not for citation purposes)
model, approximating it to the known behavior of the
biological system under study. In this way, the user has a
range of possibilities, from a purely qualitative model to
one that is highly quantitative.
There are studies that compare the dynamical behavior of
discrete and continuous dynamical systems. Hence, it is
known that while the steady state of a Boolean model will
correspond qualitatively to an analogous steady state in a
continuous approach, the reverse is not necessarily true.
Moreover, periodic solutions in one representation may
be absent in the other [16]. This discrepancy between the
discrete and continuous models is more evident for steady
states where at least one of the nodes has an activation
state precisely at, or near, its threshold of activation.
Because of this characteristic, discrete and continuous
models for a given regulatory network differ in the total
number of steady states [17]. For this reason, our method
focuses on the study of only one type of steady state;
namely, the regular stationary points [18]. These points
do not have variables near an activation threshold, and
they are always stable steady states. Moreover, it has been
shown that this type of stable steady state can be found in
discrete models, and then used to locate their analogous
states in continuous models of a given genetic regulatory
network [19].
It is beyond the scope of this paper to present a detailed
mathematical analysis of the dynamical system described
by Equation 3. Instead, we present a framework that can
help to speed up the analysis of the qualitative behavior
of signaling networks. Under this perspective, the useful-
ness of our method will ultimately be determined through
building and analyzing concrete models. To show the
capabilities of our proposed methodology, we applied it
to analysis of the regulatory network that controls differ-
entiation in T helper cells. This biological system was well
suited to evaluating our methodology because the net-
work contains several known components, and it has
three alternative stable patterns of activation. Moreover, it
is of great interest to understand the behavior of this net-
work, given the role of T helper cell subsets in immunity
and pathology [20]. Our method applied to the Th net-
work generated a model with the same qualitative behav-
ior as the biological system. Specifically, the model has
three stable states of activation, which can be interpreted
as the states of activation found in Th0, Th1 and Th2 cells.
In addition, the system is capable of being moved from
the Th0 state to either the Th1 or Th2 states, given a suffi-
ciently large IFN-γ or IL-4 signal, respectively. This charac-
teristic reflects the known qualitative properties of IFN-γ
and IL-4 as key cytokines that control the fate of T helper
cell differentiation.
Regarding the numerical values returned by the model, it
is not possible yet to evaluate their accuracy, given that (to
our knowledge) no quantitative experimental data are
available for this biological system. The resulting model,
then, should be considered as a qualitative representation
of the system. However, representing the nodes in the net-
work as normalized continuous variables will eventually
permit an easy comparison with quantitative experimen-
tal data whenever they become available. Towards this
end, the equations in our methodology define a sigmoid
function, with values ranging from 0 to 1, regardless of the
values of assigned to the parameters in the equations. This
characteristic has been used before to represent and
model the response of signaling pathways [21,22]. It is
important to note, however, that the modification of the
parameters allow the model to be fitted against experi-
mental data.
One benefit of a mathematical model of a particular bio-
logical network is the possibility of predicting the behav-
Table 5: Stable steady states of the signaling network in Figure 6
Discrete state
1
Discrete state
2
Discrete state
3
Discrete state
4
Continuous
state 1
Continuous
state 2
Continuous
state 3
Continuous
state 4
GATA3
0
0
11
1
0
0
0.93037
0.93037
IFN-γ
0
1
0
1
0
0.99914
0
0.90967
IFN-γR
0
1
0
1
0
0.99997
0
0.99617
IL-12
0
0
0
0
0
0
0
0
IL-12R
0
1
0
0
0
0.9096
0
0.00193
IL-13
0
0
1
1
0
0
0.99719
0.99719
IL-4
0
0
1
1
0
0
0.99719
0.99719
IL-4R
0
0
1
1
0
0
0.99991
0.99991
IL-5
0
0
1
1
0
0
0.99719
0.99719
STAT1
0
1
0
1
0
1
0
0.99988
STAT4
0
1
0
0
0
0.99617
0
2.4E-4
STAT6
0
0
1
1
0
0
1
1
T-bet
0
1
0
1
0
0.93037
0
0.93034
TCR
0
0
0
0
0
0
0
0
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 11 of 18
(page number not for citation purposes)
ior of complex experimental setups. Therefore, it is
important to be aware of its limitations beforehand, to
avoid generating experimental data that cannot be han-
dled by the model. The method we present in this paper
has been developed to obtain the number and relative
position of the stable steady states of a regulatory network.
Equations 1 and 3 include a number of parameters that
allow the response of the model to be fine-tuned, but the
equations were not designed to describe the transitory
responses of molecules with great detail. Therefore, failure
to predict a stable steady state with high numerical accu-
racy should not be interpreted as a failure of the approach
presented here. By contrast, failure to describe and/or pre-
dict the number and approximate location of stable
steady states under a wide range of values for the parame-
ters would call the validity of the reconstruction of a par-
ticular network into question. Here, however, it is
essential to establish the validity of the network used as
input. Indeed, we applied our method to four alternative
forms of the network that regulates Th cell differentiation.
The alternative networks (Figures 4 through 7) were taken
from previously published attempts to discover the
molecular basis of this differentiation process. Originally,
such networks were not developed with the idea of study-
ing dynamical properties. It is not surprising, then, that
these networks do not reflect the existence of three stable
steady states, representing the molecular states of Th0,
Th1 and Th2 cells, respectively. In these cases, the failure
to find the correct stable steady states is not a problem in
the modeling methodology, but a problem in the infer-
ence of the regulatory network.
In conclusion, we have shown that the creation of a
dynamical model of a regulatory network can be consid-
erably simplified with the aid of a standardized set of
equations, where the feature that distinguishes one mole-
cule from another is the number of regulatory inputs.
Such standardization permits a continuous dynamical
system to be systematically and analytically constructed
together with a basic analysis of its global properties,
based exclusively on the information provided by the con-
nectivity of the network. While the use of a standardized
set of functions to model a network may severely restrict
the capability to fit specific datasets, we believe that the
loss in flexibility is balanced by the possibility of rapidly
developing models and gaining knowledge of the dynam-
ical behavior of a network, especially in those cases where
few kinetic data are available. Thus, we provide a method
for incorporating the dynamical perspective in the analy-
sis of regulatory networks, using the topological informa-
Table 6: Stable steady states of the signaling network in Figure 7
Discrete state 1
Discrete state 2
Continuous state 1
Continuous state 2
Ag/MHC
0
0
0
0
ATF2
0
0
0
0
c-Maf
0
0
0
0
CD4
0
0
0
0
GATA3
0
1
0
0.99999
IFN-γ
0
0
0
0
IL-12
0
0
0
0
IL-12R
0
0
0
0
IL-13
0
1
0
0.8468
IL-18
0
0
0
0
IL-18R
0
0
0
0
IL-4
0
1
0
0.8468
IL-4R
0
1
0
0.99176
IL-5
0
1
0
0.8469
IRAK
0
0
0
0
Itk
0
0
0
0
JNK
0
0
0
0
JNK2
0
0
0
0
Lck
0
0
0
0
MKK3
0
0
0
0
NFAT
0
0
0
0
NFkB
0
0
0
0
p38/MAPK
0
0
0
0
STAT4
0
0
0
0
STAT6
0
1
0
0.99975
T-bet
0
0
0
0
TCR
0
0
0
0
TRAF6
0
0
0
0
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 12 of 18
(page number not for citation purposes)
tion of a network, without the need to collect extensive
time-series or kinetic data.
Methods
Molecular basis of the Th network topology
The following paragraphs detail the evidence used to infer
the topology of the Th regulatory network, updating the
data summarized in [11]. Th1 cells are producers of IFN-γ
[10,23], which acts on its target cells by binding to a cell-
membrane receptor [24-26] to start a signaling cascade,
which involves JAK1 and STAT-1 [27-29]. STAT-1 can be
activated by a number of ligands besides IFN-γ, but
importantly, it cannot be activated by IL-4 [30], which is
a major Th2 signal. In contrast, STAT-1 plays a role in
modulating IL-4, being an intermediate in the negative
regulation of IFN-γ exerted on IL-4 expression [31]. Differ-
ent signals converge in STAT-1, among them that of IFN-
β/IFN-βR [32]. The IFN-γ signaling continues downstream
to activate SOCS-1 in a STAT-1-dependent pathway
[33,34]. SOCS-1, in turn, influences both the IFN-γ and
IL-4 pathways. On the one hand, SOCS-1 is a negative reg-
ulator of IFN-γ signaling, blocking the interaction of IFN-
γR and STAT-1 [35] due to direct inhibition of JAK1
[29,36]. On the other hand, SOCS-1 blocks the IL-4R/
STAT-6 pathway [37]. SOCS-1 is, therefore, a key element
for the inhibition from the IFN-γ to the IL-4 pathway. Th1
cells express high levels of SOCS-1 mRNA, while it is
barely detectable in Th0 and Th2 cells [38]. Finally,
another key molecule is T-bet, which is a transcription fac-
tor detected in Th1 but not Th0 or Th2 cells. T-bet expres-
sion is upregulated by IFN-γ in a STAT-1-dependent
mechanism [39]. Importantly, T-bet is an inhibitor of
GATA-3 [40], an activator of IFN-γ [40] and activator of T-
bet itself [41,42].
Th2 cells express IL-4, which is the major known determi-
nant of the Th2 phenotype itself [43]. IL-4 binds to its
receptor, IL-4R, which is preferentially expressed in Th2
cells [23,44]. The IL-4R signaling is transduced by STAT-6,
which in turn activates GATA-3 [10]. GATA-3, in turn, is
capable of inducing IL-4 [45], thus establishing a feedback
loop. The influence from the IL-4 pathway on the IFN-γ
pathway seems to be mediated by GATA-3 via STAT-4
[46]. Like T-bet, GATA-3 also presents a self-activation
loop [47-49].
IL-12 and IL-18 are two molecules that affect the IFN-γ
pathway. IL-12 is a cytokine produced by monocytes and
dendritic cells and promotes the development of Th1 cells
[50]. The IL-12 receptor is present in its functional form in
Th0 and Th1 but not Th2 cells [51]. IL-12R signaling is
mediated by STAT-4 [52], which is able to activate IFN-γ
Table 7: Circuits of the Th network a
1
IFNγ→IFNγR→JAK1→STAT1¬IL4→IL4R→STAT6¬IL18R→IRAK→
2
IFNγ→IFNγR→JAK1→STAT1¬IL4→IL4R→STAT6¬IL12R→STAT4→
3
IFNγ→IFNγR→JAK1→STAT1¬IL4→IL4R→STAT6→GATA3→IL10→IL10R→STAT3¬
4
IFNγ→IFNγR→JAK1→STAT1¬IL4→IL4R→STAT6→GATA3¬STAT4→
5
IFNγ→IFNγR→JAK1→STAT1¬IL4→IL4R→STAT6→GATA3¬Tbet→
6
IFNγ→IFNγR→JAK1→STAT1→SOCS1¬IL4R→STAT6¬IL18R→IRAK→
7
IFNγ→IFNγR→JAK1→STAT1→SOCS1¬IL4R→STAT6¬IL12R→STAT4→
8
IFNγ→IFNγR→JAK1→STAT1→SOCS1¬IL4R→STAT6→GATA3→IL10→IL10R→STAT3¬
9
IFNγ→IFNγR→JAK1→STAT1→SOCS1¬IL4R→STAT6→GATA3¬STAT4→
10
IFNγ→IFNγR→JAK1→STAT1→SOCS1¬IL4R→STAT6→GATA3¬Tbet→
11
IFNγ→IFNγR→JAK1→STAT1→Tbet→
12
IFNγ→IFNγR→JAK1→STAT1→Tbet→SOCS1¬IL4R→STAT6¬IL18R→IRAK→
13
IFNγ→IFNγR→JAK1→STAT1→Tbet→SOCS1¬IL4R→STAT6¬IL12R→STAT4→
14
IFNγ→IFNγR→JAK1→STAT1→Tbet→SOCS1¬IL4R→STAT6→GATA3→IL10→IL10R→STAT3¬
15
IFNγ→IFNγR→JAK1→STAT1→Tbet→SOCS1¬IL4R→STAT6→GATA3¬STAT4→
16
IFNγ→IFNγR→JAK1→STAT1→Tbet¬GATA3→IL4→IL4R→STAT6¬IL18R→IRAK→
17
IFNγ→IFNγR→JAK1→STAT1→Tbet¬GATA3→IL4→IL4R→STAT6¬IL12R→STAT4→
18
IFNγ→IFNγR→JAK1→STAT1→Tbet¬GATA3→IL10→IL10R→STAT3¬
19
IFNγ→IFNγR→JAK1→STAT1→Tbet¬GATA3¬STAT4→
20
IL4→IL4R→STAT6→GATA3→
21
IL4R→STAT6→GATA3¬ Tbet→SOCS1¬
22
Tbet→
23
Tbet¬GATA3¬
24
GATA3→
25
IL4→IL4R→STAT6→GATA3¬Tbet→SOCS1¬JAK1→STAT1¬
26
JAK1→STAT1→SOCS1¬
27
JAK1→STAT1→Tbet→ SOCS1¬
a. If the circuit has zero or an even number of negative interactions, it is considered positive; otherwise the circuit is negative. Circuits 1–24 are
positive, and circuits 25–27 are negative.
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 13 of 18
(page number not for citation purposes)
[41,46,53]. The IL-12 signaling pathway can be blocked
by IL-4 by the STAT-6 dependent down-regulation of one
subunit of IL-12R [54]. IL-18 is a cytokine produced by
many cell types and promotes IFN-γ production in Th
cells [55]. It acts upon binding to its receptor, IL-18R,
which acts through IRAK [56]. IL-12 and IL-18 act syner-
gistically to increase IFN-γ production, but using different
pathways [57,58]. Finally, IL-4 is able to block IL-18 sign-
aling in a STAT-6 dependent manner [59].
IL-10 is a cytokine actively produced by Th2 cells, and it
inhibits cytokine production by Th1 cells. As with the
other cytokines mentioned above, IL-10 acts upon bind-
ing to a cell surface receptor, IL-10R, which in turn acti-
vates the STAT signaling system [60]. In particular, it has
been shown that the functioning of IL-10 signaling is
dependent upon the presence of STAT-3 [61]. As for the
signals affecting IL-10 expression, it has been shown that
IL-4 enhances IL-10 gene expression in Th2 but not Th1
cells [62]. This requirement implies that the intracellular
signaling from IL-4 to IL-10 should pass through a Th2
specific molecule, which from the molecules considered
here can only be GATA-3. Finally, IL-10 has been shown
to be a very powerful inhibitor of IFN-γ production
[60,63].
Cytokine gene expression in T cells is induced by the acti-
vation of the T cell receptor (TCR) by ligand binding. Dif-
ferent signaling pathways are activated by the TCR [64].
Among these is the pathway including the NFAT family of
transcription factors, which are implicated in the T cell
activation-dependent regulation of numerous cytokines.
A constitutively active form of one of the NFAT proteins,
specifically NFATc1, increases the expression of IFN-γ
[65]. Importantly, the same experimental procedure does
not affect the expression of IL-4. All this indicates that the
NFAT family members play a central role in the TCR-
Activation of a node as a function of one positive input
Figure 10
Activation of a node as a function of one positive
input. The activation of a node in response to one positive
input, plotted for various possible interaction weights.
total activation
xa
Activation of a node as a function of its total input, ω
Figure 8
Activation of a node as a function of its total input, ω.
Equation 3 ensures that the activation of a node has the form
of a sigmoid, bounded in the interval [0,1] regardless of the
values of h.
total activation
ω
Total input to a node, ω, as a function of one positive input,
xaFigure 9
Total input to a node, ω, as a function of one positive
input, xa. The value of ω is a bounded function in the inter-
val [0,1] regardless of the interaction weight of the positive
input, α.
ω
xa
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 14 of 18
(page number not for citation purposes)
induced expression of cytokines during Th cell differenti-
ation,
especially
in
the
Th1
pathway.
The discrete dynamical system
The discrete system represents the network as a series of
interconnected elements that have only two possible
states of activation, 0 (or inactive) and 1 (or active). Given
this property, the network is completely described by the
following set of Boolean equations:
Equation 1.
A node x in the network can have only one of three possi-
ble forms depending on whether it has activator and
inhibitor input nodes, or only activators, or only inhibi-
tors. In the first case, i.e. form § in Eqn.1, the Boolean
function can be read as: x will be active in the next time
step if at this time any of its activators and none of its
inhibitors are acting upon it. Similarly, form §§ can be
translated as: x will be active if any of its activators is acting
upon it. And finally, form §§§ reads as: x will be active if
none of its inhibitors are acting upon it. Note than in all
cases inhibitors are strong enough to change the state of a
node from 1 to 0, while activators are strong enough to
change the state of a node from 0 to 1 if no inhibitor is act-
ing on the node of reference. The three alternative forms
of representing a node in Equation 1 imply two possible
default states of activation, i.e. the state of a node when
there are neither activators nor inhibitors acting upon it.
If the connectivity of the node includes either only posi-
tive inputs, or both positive and negative inputs, then the
node has an inactive state by default. Alternatively, if the
connectivity of a node has only negative inputs, then the
node has an active state by default.
The Th network (Figure 2) can be converted into a discrete
dynamical system using Equation 1. The resulting system
of equations is as follows:
Equation 2.
GATA3(t + 1) = (GATA3(t) ∨ STAT6(t)) ∧ ¬(T - bet(t))
IFN - βR(t + 1) = IFN - β(t)
IFN - γ(t + 1) = (IRAK(t) ∨ NFAT(t) ∨ STAT - 4(t) ∨ T -
bet(t)) ∧ ¬(STAT3(t))
Equation 1.
x t
x
t
x
t
x
t
x t
x
t
i
a
a
n
a
i
i
(
)
( )
( )
( )
(
( )
( )
+
=
∨
∨
(
) ∧¬
∨
1
1
2
1
2
…
…
…
…
∨
∨
∨
¬
∨
∨
x
t
t
x
t
x
t
t
x
t
x
t
m
i
a
n
a
i
m
i
( ))
( )
( )
( )
( )
( )
( ))
§
x
§§
x
1
a
1
i
2
2
(
§§§
∨∧
¬
, , and are the logical operators OR, AND, and NOT
is the set of activators of
i
x
x
x
x
i
n
a
i
m
i
∈{ , }
{
}
{
}
0 1
s the set of inhibitors of
is used if
has activato
xi
§
xi
rs and inhibitors
is used if
has only activators
§§
x
§§§
i
is used if
has only inhibitors
xi
Activation of a node as a function of one negative input
Figure 12
Activation of a node as a function of one negative
input. The activation of a node in response to one negative
input, plotted for various possible interaction weights.
total activation
xi
Total input to a node, ω, as a function of one negative input,
xiFigure 11
Total input to a node, ω, as a function of one negative
input, xi. The value of ω is a bounded function in the interval
[0,1] regardless of the interaction weight of the negative
input, β.
ω
xi
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 15 of 18
(page number not for citation purposes)
IFN - γR(t + 1) = IFN - γ(t)
IL - 10(t + 1) = GATA3(t)
IL - 10R(t + 1) = IL - 10(t)
IL - 12R(t + 1) = IL - 12(t)
IL - 18R(t + 1) = IL - 18(t) ∧ ¬(STAT6(t))
IL - 4(t + 1) = GATA3(t) ∧ ¬(STAT1(t))
IL - 4R(t + 1) = IL - 4(t) ∧ ¬(SOCS1(t))
IRAK(t + 1) = IL - 18R(t)
JAK1(t + 1) = IFN - γR(t) ∧ ¬(SOCS1(t))
NFAT(t + 1) = TCR(t)
SOCS1(t + 1) = STAT1(t) ∨ T - bet(t)
STAT1(t + 1) = IFN - βR(t) ∨ JAK1(t)
STAT3(t + 1) = IL - 10R(t)
STAT4(t + 1) = IL - 12R(t) ∧ ¬(GATA3(t))
STAT6(t + 1) = IL - 4R(t)
T - bet(t + 1) = (STAT1(t) ∨ T - bet(t)) ∧ ¬(GATA3(t))
Notice that there are only 19 equations out of a total of 23
elements in the Th network. The reason is that four ele-
ments, namely IFN-β, IL-12, IL-18 and TCR, do not have
inputs. These four elements are thus treated as constants,
since there are no interactions that regulate their behavior.
Throughout the text, these four elements are considered as
having a value of 0.
Stable steady states of the discrete system
The discrete dynamical system defined by Equation 2 can
be solved in different ways to find its attractors, depend-
ing on how to update the vector state X(t) to its successor,
X(t+1). By far the easiest method for solving the equations
is the synchronous approach (as in [66,67]). This method,
however, can generate spurious results (see [14]). Hence,
we use generalized logical analysis to find all the steady
states of the system [15]. Generalized logical analysis
allows us to find all the steady states of a discrete dynam-
ical system by evaluating the functionality of the feedback
loops, also known as circuits, in the system. In this case,
the Th network (Figure 2) contains a total of 27 circuits
(Table 7), 24 positive and 3 negative. Depending on the
set of parameters used, positive feedback loops can gener-
ate multistationarity, while negative feedback loops can
generate damped or sustained oscillations. Generalized
logical analysis is a well-established method and the
reader may find in-depth explanations elsewhere
[14,15,18].
The continuous dynamical system
To describe the network as a continuous dynamical sys-
tem, we use the following set of ordinary differential
equations:
Equation 3.
The right-hand side of the differential equation comprises
two parts: an activation function and a term for decay.
Activation is a sigmoid function of ω, which represents the
total input to the node. The equation of the sigmoid was
chosen so as to pass through the two points (0,0) and
(1,1), regardless of the value of its gain, h; see Figure 8. The
bounding of a node x to the closed interval [0,1] implies
that its level of activation should be interpreted as a nor-
malized, not an absolute, value. This characteristic permits
direct comparison between the discrete and the continu-
ous dynamical systems, since in both formalisms the min-
imum and maximum levels of activation are 0 and 1.
Subsequently, the second part of the equation is a decay
term, which for simplicity is directly proportional to the
level of activation of the node.
The total input to a node, represented by ω, is a combina-
tion of the multiple activatory and inhibitory interactions
acting upon the node of reference. In the general case, dif-
ferent nodes have different connectivities; hence it is nec-
essary to write a function ω so that it can describe different
combinations of activatory and inhibitory inputs. For this
Equation 3.
dx
dt
e
e
e
e
i
h
h
h
h
i
i
=
−
+
−
+
−
−
−
−
0 5
0 5
0 5
0 5
1
1
.
(
. )
.
(
. )
(
)(
ω
ω
)
−
=
+
+
−
+
∑
∑
∑
∑
∑
∑
γ
ω
α
α
α
α
β
β
i i
i
n
n
n n
a
n n
a
m
m
x
x
x
1
1
1
1
+
+
∑
∑
∑
∑
β
β
α
α
α
m m
i
m m
i
n
n
n
x
x
1
1
§
x
x
x
x
n
a
n n
a
m
m
m m
i
m m
i
∑
∑
∑
∑
∑
∑
+
−
+
+
1
1
1
1
α
β
β
β
β
§§
≤
≤
≤
≤
>
§§§
0
1
0
1
0
x
h
x
i
i
n
m
i
n
a
ω
α
β
γ
,
,
,
{
}
{
}
is the set of activators of
is the set of inhib
x
x
i
n
i
itors of
is used if
has activators and inhibitors
xi
§
x
§
i
§
x
§§§
x
i
i
is used if
has only activators
is used if
has only inhibitors
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 16 of 18
(page number not for citation purposes)
reason, ω has three possible forms in Equation 3. If a node
xi is regulated by both activators and inhibitors, then the
first form, §, is used. However, if is regulated exclusively
by activators, form §§ is used instead. Finally, the form
§§§ is used if xi has only negative regulators. In all cases,
the total input is a combination of weighted activators
and/or inhibitors, where the weights are represented by
the α and β parameters for the activators and inhibitors,
respectively. The mathematical form ω was chosen so as to
be monotonic and to be bounded in the closed interval
[0,1] given that 0≤x≤1, α>0 and β>0. Figure 9 shows the
behavior of ω when a node is controlled only by one acti-
vator. Notice that regardless of the value of α, the function
is monotonically increasing and bounded to [0,1]. The
reason for choosing a monotonic bounded function for ω
is to preserve the sigmoid form of the total activation act-
ing upon a node xi, irrespective of the number and nature
of the regulatory inputs acting upon it. Indeed, Figure 10
shows the total activation of a node xi controlled by one
positive regulation with different weights. Notice that the
total activation retains a bounded sigmoid form inde-
pendently of the value of α. This same qualitative behav-
ior for total activation on a node xi is observed if it is
regulated only by inhibitors. Figure 11 shows ω as a func-
tion of one inhibitor, plotted for different strengths of
interaction. In this case, the total input to xi is still a
bounded sigmoid regardless of the value of the parameter
β (see Figure 12). This general qualitative behavior per-
sists even with a mixture of activatory and inhibitory
inputs acting upon a node. Figure 13 presents the total
activation of a node xi as a function of two regulatory
inputs, one positive and one negative. Notice again that
the equation warrants a bounded sigmoid form for the
total input to a node.
Once a network is translated to a dynamical system using
Equation 3, it is necessary to specify values for all param-
eters. For a system with n nodes and m interactions, there
are m+2n parameters. However, there are usually insuffi-
cient experimental data to assign realistic values for each
and every one of the parameters. Nevertheless, it is possi-
ble to use a series of default values for all the parameters
in Equation 3. The reason is that, as we showed in the pre-
vious paragraph, the equations have the same qualitative
shape for any value assigned to the parameters. Hence, for
the sake of simplicity, it is possible to assign the same val-
ues to most of the parameters, as a first approach. For the
present study on the Th model, we use a value of 1 for all
αs, βs and γs; and we use h = 10, since we currently lack
quantitative data to estimate more realistic values. More-
over, the use of default values ensures the possibility of
creating the dynamical system in a fully automated way.
Nonetheless, after the initial construction and analysis of
the resulting system, the modeler may modify the values
of the parameters so as to fine-tune the dynamical behav-
ior of the equations, whenever more experimental quanti-
tative data become available. The continuous dynamical
system of the Th model, constructed with the use of Equa-
tion 3, yields a system of 23 equations, which is included
in the file "Th_continuous_model.octave.txt".
Stable steady states of the continuous system
Nonlinear systems of ordinary differential equations are
studied numerically. Hence the continuous dynamical
system defined by Equation 3 poses the problem of how
to find all its stable steady states without using very time-
consuming and computing-intensive methods. This is
where the creation of two dynamical systems of the same
network, one discrete and one continuous, bears fruit.
Since a Boolean (step) function is a limiting case of a very
steep sigmoid curve, networks made of binary elements
share many qualitative features with systems modeled
using continuous functions [68]. Indeed, it has been
shown [19] that the qualitative information resulted from
generalized logical analysis can be directly used to find the
number, nature and approximate location of the steady
states of a system of differential equations representing
the same network. We therefore decided to use this char-
acteristic to speed up the process of finding all the stable
steady states in the continuous dynamical system. Specif-
ically, the stable steady states of the discrete system are
used as initial states to solve the differential equations,
running them until the system converges to its own stable
steady states. Calculating the convergence of a system of
ordinary differential equations from a given initial state is
a straightforward procedure using any numerical solver.
Activation of a node as a function two inputs, one positive
and one negative
Figure 13
Activation of a node as a function two inputs, one
positive and one negative. The strength of the interac-
tions are equal for the activation and the inhibition, α = β =
1.
xi
xa
total activation
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 17 of 18
(page number not for citation purposes)
For our simulations we used the lsode function of the GNU
Octave package http://www.octave.org, stopping the
numerical integration when all the variables of the system
changed by less than 10-4 for at least 10 consecutive steps
of the procedure. The final values of the variables in the
system are considered to be the stable steady states of the
continuous model of the network.
Implementation
The methodology was fully implemented in a java pro-
gram, and it has been tested under a linux environment
using java version 1.5.0 (JRE 5.0), as well as octave version
2.1.34. The bytecode version of the program is included as
Additional file 2.
Competing interests
The author(s) declare that they have no competing inter-
ests.
Authors' contributions
LM inferred the regulatory network, created the equations,
developed the methods and wrote the paper. IX made a
substantial contribution to the design and development
of the methods, revised the intellectual content, and
helped in drafting the manuscript.
Additional material
Acknowledgements
We want to thank Massimo de Francesco, Mark Ibberson, Caroline John-
son-Leger, Maria Karmirantzou, Lukasz Salwinski, François Talabot and
Francisca Zanoguera for their valuable comments and suggestions.
References
1.
Jeong H, Tombor B, Albert R, Oltvai ZN, Barabási AL: The large-
scale organization of metabolic networks. Nature 2000,
407:651-654.
2.
Milo R, Shen-Orr S, Itzkovitz S, Kashtan N, Chklovskii D, Alon U:
Network motifs: Simple building blocks of complex net-
works. Science 2002, 298:824-827.
3.
Ravasz E, Somera AL, Mongru DA, Oltvai ZN, Barabási AL: Hierar-
chical organization of modularity in metabolic networks. Sci-
ence 2002, 297:1551-1555.
4.
Covert MW, Schilling CH, Famili I, Edwards JS, Goryanin II, Selkov E,
Palsson BO: Metabolic modeling of microbial strains in silico.
Trends Biochem Sci 2001, 26:179-186.
5.
Herrgård MJ, Covert MW, Palsson BØ: Reconstruction of micro-
bial transcriptional regulatory networks. Curr Opin Biotechnol
2004, 15:70-77.
6.
Mendoza L, Thieffry D, Alvarez-Buylla ER: Genetic control of
flower morphogenesis in Arabidopsis thaliana: a logical analy-
sis. Bioinformatics 1999, 15:593-606.
7.
Sánchez L, Thieffry D: Segmenting the fly embryo: a logical
analysis of the pair-rule cross-regulatory module. J Theor Biol
2003, 224:517-537.
8.
de Jong H: Modeling and simulation of genetic regulatory sys-
tems: a literature review. J Comp Biol 2002, 9:67-103.
9.
Lok L, Brent R: Automatic generation of cellular reaction net-
works with Moleculizer 1.0. Nature Biotechnol 2005, 23:131-136.
10.
Murphy KM, Reiner SL: The lineage decisions on helper T cells.
Nat Rev Immunol 2002, 2:933-944.
11.
Mendoza L: A network model for the control of the differenti-
ation process in Th cells. BioSystems in press.
12.
Remy E, Ruet P, Mendoza L, Thieffry D, Chaouiya C: From Logical
Regulatory Graphs to Standard Petri Nets: Dynamic Roles
and Functionality of Feedback Circuits. Transactions on Compu-
tational Systems Biology in press.
13.
Mendes P: Biochemistry by numbers: simulation of biochemi-
cal pathways with Gepasi 3. Trends Biochem Sci 1997, 22:361-363.
14.
Thomas R: Regulatory networks seen as asynchronous autom-
ata: a logical description. J Theor Biol 1991, 153:1-23.
15.
Thomas R, Thieffry D, Kaufman M: Dynamical behaviour of bio-
logical regulatory networks-I. Biological role of feedback
loops and practical use of the concept of the loop-character-
istic state. Bull Math Biol 1995, 57:247-276.
16.
Glass L, Kaufman M: The logical analysis of continuous, non-lin-
ear biochemical control networks. J Theor Biol 1973,
39:103-129.
17.
Mochizuki A: An analytical study of the number of steady
states in gene regulatory networks. J Theor Biol 2005,
236:291-310.
18.
Snoussi EH, Thomas R: Logical identification of all steady states:
the concept of feedback loop characteristic states. Bull Math
Biol 1993, 55:973-991.
19.
Muraille E, Thieffry D, Leo O, Kaufman M: Toxicity and neuroen-
docrine regulation of the immune response: a model analy-
sis. J Theor Biol 1996, 183:285-305.
20.
Singh VK, Mehrotra S, Agarwal SS: The paradigm of Th1 and Th2
cytokines: its relevance to autoimmunity and allergy. Immu-
nol Res 1999, 20:147-161.
21.
Hautaniemi S, Kharait S, Iwabu A, Wells A, Lauffenburger DA: Mod-
eling of signal-response cascades using decision tree analysis.
Bioinformatics 2005, 21:2027-2035.
22.
Sachs K, Perez O, Pe'er D, Lauffenburger DA, Nolan GP: Causal
protein-signaling networks derived from multiparameter
single-cell data. Science 2005, 308:523-529.
23.
Hamalainen H, Zhou H, Chou W, Hashizume H, Heller R, Lahesmaa
R: Distinct gene expression profiles of human type 1 and type
2 T helper cells. Genome Biology 2001, 2:1-0022.
24.
Groux H, Sornasse T, Cottrez F, de Vries JE, Coffman RL, Roncarolo
MG, Yssel H: Induction of human T helper cell type 1 differen-
tiation results in loss of IFN-γ receptor β-chain expression. J
Immunol 1997, 158:5627-5631.
25.
Novelli F, D'Elios MM, Bernabei P, Ozmen L, Rigamonti L, Almeri-
gogna F, Forni G, Del Prete G: Expression and role in apoptosis
of the α- and β-chains of the IFN-γ receptor in human Th1
and Th2 clones. J Immunol 1997, 159:206-213.
26.
Rigamonti L, Ariotti S, Losana G, Gradini R, Russo MA, Jouanguy E,
Casanova JL, Forni G, Novelli F: Surface expression of the IFN-
γR2 chain is regulated by intracellular trafficking in human T
lymphocytes. J Immunol 2000, 164:201-207.
Additional File 1
The file contains the set of differential equations describing the continuous
version of the Th model. It is a plain text file formatted for running sim-
ulations using the GNU Octave package http://www.octave.org
Click here for file
[http://www.biomedcentral.com/content/supplementary/1742-
4682-3-13-S1.txt]
Additional File 2
The file is a java program that implements the methodology described in
this paper; it requires a working installation of GNU Octave http://
www.octave.org. The program takes as input a plain text file containing
the topology of the network to analyze, with the following format: Mole-
culeA -> MoleculeB MoleculeB -| MoleculeA The output of the program is
a stream of plain text formatted for GNU Octave.
Click here for file
[http://www.biomedcentral.com/content/supplementary/1742-
4682-3-13-S2.jar]
Theoretical Biology and Medical Modelling 2006, 3:13
http://www.tbiomed.com/content/3/1/13
Page 18 of 18
(page number not for citation purposes)
27.
Kotenko SV, Pestka S: Jak-Stat signal transduction pathway
through the eyes of cytokine class II receptor complexes.
Oncogene 2000, 19:2557-2565.
28.
Kerr IM, Costa-Pereira AP, Lillemeier BF, Strobl B: Of JAKs,
STATs, blind watchmakers, jeeps and trains. FEBS Lett 2003,
546:1-5.
29.
Krebs DL, Hilton DJ: SOCS proteins: negative regulators of
cytokine signaling. Stem Cells 2001, 19:378-387.
30.
Moriggl R, Kristofic C, Kinzel B, Volarevic S, Groner B, Brinkmann V:
Activation of STAT proteins and cytokine genes in human
Th1 and Th2 cells generated in the absence of IL-12 and IL-
4. J Immunol 1998, 160:3385-3392.
31.
Elser B, Lohoff M, Kock S, Giaisi M, Kirchhoff S, Krammer PH, Li-
Weber M: IFN-γ represses IL-4 expression via IRF-1 and IRF-
2. Immunity 2002, 17:703-712.
32.
Goodbourn S, Didcock L, Randal RE: Interferons: cell signalling,
immune modulation, antiviral responses and virus counter-
measures. J Gen Virol 2000, 81:2341-2364.
33.
Chen XP, Losman JA, Rothman P: SOCS proteins, regulators of
intracellular signaling. Immunity 2000, 13:287-290.
34.
Saito H, Morita Y, Fujimoto M, Narazaki M, Naka T, Kishimoto T:
IFN regulatory factor-1-mediated transcriptional activation
of mouse STAT-induced STAT inhibitor-1 gene promoter
by IFN-γ. J Immunol 2000, 164:5833-5843.
35.
Diehl S, Anguita J, Hoffmeyer A, Zapton T, Ihle JN, Fikrig E, Rincón M:
Inhibition of Th1 differentiation by IL-6 is mediated by
SOCS1. Immunity 2000, 13:805-815.
36.
Zhang JG, Metcalf D, Rakar S, Asimakis M, Greenhalgh CJ, Willson
TA, Starr R, Nicholson SE, Carter W, Alexander WS, Hilton J: The
SOCS box of suppressor of cytokine signaling-1 is important
for inhibition of cytokine action in vivo. Proc Natl Acad Sci USA
2001, 98:13261-13265.
37.
Losman JA, Chen XP, Hilton D, Rothman P: Cutting edge: SOCS-
1 is a potent inhibitor of IL-4 signal transduction. J Immunol
1999, 162:3770-3774.
38.
Egwuagu CE, Yu CR, Zhang M, Mahdi RM, Kim SJ, Gery I: Suppres-
sors of cytokine signaling proteins are differentially
expresses in Th1 and Th2 cells: implications for the Th cell
lineage commitment and maintenance. J Immunol 2002,
168:3181-3187.
39.
Lighvani A., Frucht DM, Jankovic D, Yamane H, Aliberti J, Hissong BD,
Nguyen BV, Gadina M, Sher A, Paul WE, O'Shea JJ: T-bet is rapidly
induced by interferon-γ in lymphoid and myeloid cells. Proc
Natl Acad Sci USA 2001, 98:15137-15142.
40.
Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH: A
novel transcription factor, T-bet, directs Th1 lineage com-
mitment. Cell 2000, 100:655-669.
41.
Mullen AC, High FA, Hutchins AS, Lee HW, Villarino AV, Livingston
DM, Kung AL, Cereb N, Yao TP, Yang SY, Reiner SL: Role of T-bet
in commitment of TH1 cells before IL-12-dependent selec-
tion. Science 2001, 292:1907-1910.
42.
Zhang Y, Apilado R, Coleman J, Ben-Sasson S, Tsang S, Hu-Li J, Paul
WE, Huang H: Interferon γ stabilizes the T helper cell type 1
phenotype. J Exp Med 2001, 194:165-172.
43.
Agnello D, Lankford CSR, Bream J, Morinobu A, Gadina M, O'Shea J,
Frucht DM: Cytokines and transcription factors that regulate
T helper cell differentiation: new players and new insights. J
Clin Immunol 2003, 23:147-161.
44.
Nelms K, Keegan AD, Zamorano J, Ryan JJ, Paul WE: The IL-4
receptor: signaling mechanisms and biologic functions. Annu
Rev Immunol 1999, 17:701-738.
45.
Ouyang W, Löhning M, Gao Z, Assenmacher M, Ranganath S, Rad-
bruch A, Murphy KM: Stat6-independent GATA-3 autoactiva-
tion directs IL-4-independent Th2 development and
commitment. Immunity 2000, 12:27-37.
46.
Usui T, Nishikomori R, Kitani A, Strober W: GATA-3 suppresses
Th1 development by downregulation of Stat4 and not
through effects on IL-12Rβ2 chain or T-bet. Immunity 2003,
18:415-428.
47.
Zhou M, Ouyang W, Gong Q, Katz SG, White JM, Orkin SH, Murphy
KM: Friend of GATA-1 Represses GATA-3-dependent activ-
ity in CD4+ cells. J Exp Med 2001, 194:1461-1471.
48.
Zhou M, Ouyang W: The function role of GATA-3 in Th1 and
Th2 differentiation. Immunol Res 2003, 28:25-37.
49.
Höfer T, Nathansen H, Löhning M, Radbruch A, Heinrich R: GATA-
3 transcriptional imprinting in Th2 lymphocytes: A mathe-
matical model. Proc Natl Acad Sci USA 2002, 99:9364-9368.
50.
Trinchieri G: Interleukin-12: a proinflammatory cytokine with
immunoregulatory functions that bridge innate resistance
and antigen-specific adaptive immunity. Annu Rev Immunol
1995, 13:251-276.
51.
Szabo SJ, Jacobson NG, Dighe AS, Gubler U, Murphy KM: Develop-
mental commitment to the Th2 lineage by extinction of IL-
12 signaling. Immunity 1995, 2:665-675.
52.
Thierfelder WE, van Deursen JM, Yamamoto K, Tripp RA, Sarawar
SR, Carson RT, Sangster MY, Vignali DA, Doherty PC, Grosveld GC,
Ihle JN: Requirement for Stat4 in interleukin-12-mediated
responses of natural killer and T cells. Nature 1996,
382:171-174.
53.
Kaplan MH, Sun YL, Hoey T, Grusby MJ: Impaired IL-12 responses
and enhanced development of Th2 cells in Stat4-deficient
mice. Nature 1996, 382:174-177.
54.
Szabo SJ, Dighe AS, Gubler U, Murphy KM: Regulation of the inter-
leukin (IL)-12R β2 subunit expression in developing T helper
1 (Th1) and Th2 cells. J Exp Med 1997, 185:817-824.
55.
Swain SL: Interleukin 18: tipping the balance towards a T
helper cell 1 response. J Exp Med 2001, 194:F11-F14.
56.
Chang JT, Segal BM, Nakanishi K, Okamura H, Shevach EM: The cos-
timulatory effect of IL-18 on the induction of antigen-specific
IFN-gamma production by resting T cells is IL-12 dependent
and is mediated by up-regulation of the IL-12 receptor beta2
subunit. Eur J Immunol 2000, 30:1113-1119.
57.
Akira S: The role of IL-18 in innate immunity. Curr Opin Immunol
2000, 12:59-63.
58.
Kanakaraj P, Ngo K, Wu Y, Angulo A, Ghazal P, Harris CA, Siekierka
JJ, Peterson PA, Fung-Leung WP: Defective interleukin (IL)-18-
mediated natural killer and T helper cell type 1 response in
IL-1 receptor-associated kinase (IRAK)-deficient mice. J Exp
Med 1999, 189:1129-1138.
59.
Smeltz RB, Chen J, Hu-Li J, Shevach EM: Regulation of interleukin
(IL)-18 receptor α chain expression on CD4+ T cells during
T helper (Th)1/Th2 differentiation: critical downregulatory
role of IL-4. J Exp Med 2001, 194:143-153.
60.
Moore KW, de Waal Malefyt R, Coffman R, O'Garra A: Interleukin-
10 and the interleukin-10 receptor. Annu Rev Immunol 2001,
19:683-765.
61.
Riley JK, Takeda K, Akira S, Schreiber RD: Interleukin-10 receptor
signaling through the JAK-STAT pathway. J Biol Chem 1999,
274:16513-16521.
62.
Schmidt-Weber CB, Alexander SI, Henault LE, James L, Lichtman AH:
IL-4 enhances IL-10 gene expression in murine Th2 cells in
the absence of TCR engagement. J Immunol 1999, 162:238-244.
63.
Skapenko A, Niedobitek GU, Kalden JR, Lipsky PE, Schulze-Koops H:
Generation and regulation of human Th1-biased immune
response in vivo: A critical role for IL-4 and IL-10. J Immunol
2004, 172:6427-6434.
64.
Huang Y, Wange RL: T cell receptor signaling: beyond complex
complexes. J Biol Chem 2004, 279:28827-28830.
65.
Porter CM, Clipstone NA: Sustained NFAT signaling promotes
a Th1-like pattern of gene expression in primary murine
CD4+ T cells. J Immunol 2002, 168:4936-4945.
66.
Kauffman SA: Metabolic stability and epigenesis in randomly
constructed genetic nets. J Theor Biol 1969, 22:437-467.
67.
Kauffman SA: Antichaos and adaptation. Sci Am 1991, 265:78-84.
68.
Thomas R: Laws for the dynamics of regulatory networks. Int
J Dev Biol 1998, 42:479-485.
69.
Muraille E, Leo O: Revisiting the Th1/Th2 paradigm. Scand J
Immunol 1998, 47:1-9.
70.
Street NE, Mosmann TM: Functional diversity of T lymphocytes
due to secretion of different cytokine patterns. FASEB J 1991,
5:171-177.
71.
Murphy KM, Ouyang W, Farrar JD, Yang J, Ranganath S, Asnagli H,
Afkarian M, Murphy TL: Signaling and transcription in T helper
development. Annu Rev Immunol 2000, 18:451-494.
|
16542429
|
IL4 = ( ( GATA3 ) AND NOT ( STAT1 ) )
SOCS1 = ( Tbet ) OR ( STAT1 )
IFNg = ( ( STAT4 ) AND NOT ( STAT3 ) ) OR ( ( IRAK ) AND NOT ( STAT3 ) ) OR ( ( Tbet ) AND NOT ( STAT3 ) ) OR ( ( NFAT ) AND NOT ( STAT3 ) )
STAT3 = ( IL10R )
IRAK = ( IL18R )
IFNgR = ( IFNg )
STAT1 = ( IFNbR ) OR ( JAK1 )
NFAT = ( TCR )
STAT6 = ( IL4R )
IL4R = ( ( IL4 ) AND NOT ( SOCS1 ) )
GATA3 = ( ( STAT6 ) AND NOT ( Tbet ) ) OR ( ( GATA3 ) AND NOT ( Tbet ) )
JAK1 = ( ( IFNgR ) AND NOT ( SOCS1 ) )
IL12R = ( IL12 )
IL18R = ( ( IL18 ) AND NOT ( STAT6 ) )
STAT4 = ( ( IL12R ) AND NOT ( GATA3 ) )
IL10R = ( IL10 )
IFNbR = ( IFNb )
Tbet = ( ( Tbet ) AND NOT ( GATA3 ) ) OR ( ( STAT1 ) AND NOT ( GATA3 ) )
IL10 = ( GATA3 )
|
Emergent decision-making in biological signal
transduction networks
Toma´sˇ Helikar*, John Konvalina†, Jack Heidel†, and Jim A. Rogers*†‡
*Department of Pathology and Microbiology, University of Nebraska Medical Center, 983135 Nebraska Medical Center, Omaha, NE 68198; and †Department
of Mathematics, University of Nebraska, 6001 Dodge Street, Omaha, NE 68182
Edited by Eugene V. Koonin, National Institutes of Health, Bethesda, MD, and accepted by the Editorial Board December 14, 2007 (received for review
May 30, 2007)
The complexity of biochemical intracellular signal transduction net-
works has led to speculation that the high degree of interconnectivity
that exists in these networks transforms them into an information
processing network. To test this hypothesis directly, a large scale
model was created with the logical mechanism of each node de-
scribed completely to allow simulation and dynamical analysis. Ex-
posing the network to tens of thousands of random combinations of
inputs and analyzing the combined dynamics of multiple outputs
revealed a robust system capable of clustering widely varying input
combinations into equivalence classes of biologically relevant cellular
responses. This capability was nontrivial in that the network per-
formed sharp, nonfuzzy classifications even in the face of added
noise, a hallmark of real-world decision-making.
information processing systems biology
I
ntracellular signal transduction is the process by which chemical
signals from outside the cell are passed through the cytoplasm to
cellular systems, such as the nucleus or cytoskeleton, where appro-
priate responses to those signals are generated. Unlike classical
biochemical pathways (such as those involved in various metabolic
activities) that are generally well understood and characterized by
a degree of understandability and efficiency that can be described
as elegant, signal transduction pathways are noted for their non-
linear, highly interconnected nature. Stimulation of a given cell
surface receptor can induce the activation of a network of tens or
even hundreds of cytoplasmic proteins; these networks are not
necessarily receptor-specific because different receptors, even those
associated with highly differing cellular functions, often activate
common sets of proteins (1–3). How differential responses are
generated by these networks is not obvious nor is the reason cells
evolved such a complicated mechanism for transducing signals.
Thus, a full understanding of the mechanism of intracellular signal
transduction remains a major challenge in cellular biology.
Similarities in the structure of signal transduction networks to
parallel distributed processing networks have led to speculation that
signal transduction may involve more than simple passing along of
signals. One hypothesis is that signal transduction pathways func-
tion as an information-processing system that confers nontrivial
decision-making ability (4–8). The number and variety of surface
receptors indicates that cells, either as single cells or as part of
multicellular organisms, likely encounter a large amount of infor-
mation from their environments. Thus, surface receptors function
as cellular sensory systems that bring in information that must be
centralized and integrated and the proper cellular response de-
cided. Decision-making in real-world cellular environments (which
are often chaotic, noisy, or contradictory) is unlikely to be relatively
trivial (e.g., linear feedback) but, rather, a higher-order, nontrivial
decision-making function analogous to neural networks. The ability
of individual cells to process information and make nontrivial
decisions would have an obvious advantage in terms of adaptation
but might also characterize a fundamental difference between living
and nonliving systems (9, 10).
Testing the hypothesis of signal transduction networks as non-
trivial decision-making systems requires a systems biology approach
because it is likely that the decision-making function is an emergent
property of the entire system working in concert (11–13). Numer-
ous studies have been performed on the static connectivity maps of
signal transduction networks to compare them to other naturally
occurring large-scale networks (14). The next major step to extend
these results (and a crucial requirement to test explicitly for
emergent functions multifamily signal transduction networks) is to
study the actual dynamics of a large-scale system (15). To simulate
and observe the dynamics of a system, each node’s logic (or
‘‘instruction set’’) for activation must be determined based on the
activation states of all of its regulatory inputs; i.e., the complete logic
of each node in the system must be taken into account. In this study,
we have created a large-scale model of signal transduction consist-
ing of three major receptor families; receptor tyrosine kinases
(RTKs), G protein-coupled receptors (GPCR), and Integrins.
Using logical instruction sets for each node derived from mecha-
nistic data in the biochemical literature, we show that this signal
transduction network is able to perform nontrivial pattern recog-
nition, a high-level activity associated with decision-making in
machine learning. Nontrivial pattern recognition involves decision-
making based on input information that is not necessarily clean or
clear-cut; i.e., decision-making in real-world environments. In ad-
dition to the ability to classify clearly even relatively indistinct
inputs, we show this pattern recognition function is robust in that
it is able to perform even under high noise conditions. Together,
these results are strong evidence that intracellular signal transduc-
tion networks have emergent functions that are characteristic of a
nontrivial decision-making system.
Results and Discussion
Because of the highly organized nature of the cytoplasm, the size
and shape of the kinetic curves representing the in vitro interaction
of two signal transduction elements may not represent the true
interaction of those elements in the cell. Because of this, continu-
ous, differential equation-based models are difficult to parameter-
ize realistically. This is an important limitation because the dynam-
ics of a continuous model depend highly on the parameter values
used. In cases where the function of the system being modeled is
known, it is sometimes possible to reverse-engineer the parameters
necessary for a continuous model (16–18). In the present study, the
emergent functions of the network are only hypothesized, therefore
making the reverse-engineering of parameters impossible.
To avoid the problem of parameterizing a quantitative model, a
discrete Boolean model of signal transduction was created (19, 20).
Author contributions: J.K., J.H., and J.A.R. designed research; T.H., J.K., and J.A.R. per-
formed research; T.H., J.K., and J.A.R. contributed new reagents/analytic tools; T.H., J.K.,
and J.A.R. analyzed data; and J.A.R. wrote the paper.
The authors declare no conflict of interest.
This article is a PNAS Direct Submission. E.V.K. is a guest editor invited by the Editorial
Board.
‡To whom correspondence should be addressed. E-mail: jrogers@unmc.edu.
This article contains supporting information online at www.pnas.org/cgi/content/full/
0705088105/DC1.
© 2008 by The National Academy of Sciences of the USA
www.pnas.orgcgidoi10.1073pnas.0705088105
PNAS
February 12, 2008
vol. 105
no. 6
1913–1918
CELL BIOLOGY
Because Boolean logic is qualitative in nature (21), there is no need
to consider the parameters associated with the individual protein
interactions (e.g., initial concentration, pH, etc.). The qualitative
logic of cytoplasmic protein interactions is generally straightfor-
ward to derive from the biochemical literature, where results are
usually expressed in qualitative terms (e.g., protein x activates
protein y, protein z deactivates y, etc.). Beginning with the classical
epidermal growth factor receptor (EGFR) to extracellular signal-
regulated kinase (Erk) pathway, all upstream interactions for each
member of the pathway were determined by extensive search of the
literature, and a logic table (representing an instruction set) was
created for each protein node. For each node, the logic table, the
literature cited, and an explanation of how the logic was determined
can be viewed in an online database, which can be found at
http://mathbio.unomaha.edu/Database, and further details on the
modeling can be found in Materials and Methods as well as sup-
porting information (SI) Text. It should be noted that no automated
methods were used in the creation of the database, rather, all papers
(nearly 800) were read and all pertinent information added to the
database by hand. The extent of the connectivity of the network can
be seen in Fig. 1.
To test the model’s ability to replicate known qualitative behav-
iors of the actual biological network, tests were first conducted to
find the optimal input settings to do controlled experiments. This
is directly analogous to optimization experiments in actual labora-
tory studies (e.g., determining the optimal medium and plating
conditions of a cell before performing a growth factor titration).
Thus, a sample of 10,000 random inputs was applied to the network
and the behavior of individual outputs was correlated with selected
inputs as shown in SI Text. Based on these results, optimized
conditions were determined and controlled, qualitative input–
output experiments were performed by using those conditions with
the input of interest varying from 0% to 100%. The results of those
controlled experiments can be seen in Fig. 2 and show that many
classical, input–output functional relationships in the literature are
reproduced by the model. These include the classical relationships
of each family of receptors and known interdependencies between
those families. These results indicate that Boolean logic can be used
to describe each node of a large-scale intracellular signal transduc-
tion network qualitatively and the resulting model replicates many
of the major known activities of the original system.
With a functioning, large-scale Boolean model of intracellular
signal transduction in hand, the next step was to test the hypothesis
of emergent information-processing functions in the system. This
was accomplished by applying a sample of 10,000 random, stress-
limited input combinations and categorizing the activity of the
individual output nodes by using three different ranges; 0 (0–9%
ON), 1 (10–29% ON), and 2 (30–100% ON), as shown in Fig. 1.
Based on these categories, the combined response of all four
outputs (i.e., the global response) to a given input combination can
be expressed as a ternary string of length four, with each bit
representing an individual output node. These ranges were chosen
because they reduce the global output space to a more manageable
size (34 81 states) and because ranges of this size are at the limits
of resolution of actual laboratory data commonly used (e.g.,
blotting). Results presented do not depend on these ranges because
experiments were performed with different ranges (from three to
six) with very similar results (see SI Text). These runs were
performed at 2% noise (a baseline noise level described more
fully below).
The results of this analysis with 10,000 runs is shown in Table 1.
The most striking aspect of the results shown in Table 1 is the
relatively small number of global outputs. There are 34 81
possible global outputs of the system, but after 10,000 different
inputs, only 38 outputs (50% of the total global output space) are
observed, many at low frequency. There are only 15 outputs that
Fig. 1.
The Boolean model of signal transduction and method of simulation.
The actual connection graph of the 130-node Boolean model is shown inside
the cell. The inputs are external to the cell and the outputs are nodes that are
part of the network and thus inside the cell. The four nonstress output nodes
were selected on the basis of their role in regulating other major cellular
functions, as indicated. The stress outputs are the two stress-activated protein
kinases SAPK and p38. As a demonstration of how simulations are performed,
four random inputs are applied to the network, indicated as runs 1–4. These
inputs are stress-limited because the stress inputs are limited to values be-
tween 0% and 5% ON, whereas the nonstress inputs are random values
between 0% and 100% ON. After the application of each of the inputs, the
network is iterated until it reaches a cycle, and the percentage ON of each
output is calculated. This results in four corresponding individual outputs,
shown at the bottom. The global outputs are the combination of all four
individual outputs and are represented by conversion to a ternary string
(shown on the bottom right) based on the ranges described in the text.
Fig. 2.
Qualitative, individual input–output relationships in the Boolean
model of signal transduction. (A) Positive relationship between EGF and Akt
(25). (B) Positive relationship between EGF and Erk (34). (C) EGF dependence
on Integrin stimulation by extracellular matrix (ECM) proteins for Erk stimu-
lation (27). (D) Low-level stimulation of Erk by high levels of ECM (36). (E)
Hormonal stimulators (alphaslig) of G-associated GPCR activation of adenyl-
ate cyclase (AC) (37, 38). (F) GPCR activation of Erk. (39) (G) GPCR stimulation
of Erk depends on transactivation of the EGFR (40). (H and I) Activation of the
stress-associated MAPK’s SAPK and p38 by stress (33, 34). (J and K) Activation
of Rac and Cdc42 by ECM (28). (L) Activating mutations of known protoon-
cogenes such as Ras result in growth factor-independent activation of Erk (41).
Note that the references refer to classical, qualitative input–output relation-
ships (not necessarily quantitative dose–response curves), and the dose–
response curves presented here are intended to demonstrate how the Bool-
ean model qualitatively reproduces the referenced input– output
relationships over a range of inputs.
1914
www.pnas.orgcgidoi10.1073pnas.0705088105
Helikar et al.
appear 100 times (19% of the total output space), and they
account for 9,389 (94%) of the runs. This result is even more
dramatic when the global outputs are categorized by using six
different ranges; 0–10%, 11–20%, 21–30%, 31–40%, 41–50%, and
50%. With these six ranges, the size of the output space is
64 1,296 states, yet there are only 24 outputs (1.9%) that occur
100 times, accounting for 8,597 (86%) of the inputs run (SI
Table 5).
The average degree (K) of the current network is 4.4, and the
average bias (P) is 69.8%. Although these parameters are
predictive of relatively chaotic behavior in autonomous Boolean
networks (22), the ordered behavior seen in the relatively small
number of global responses may be a reflection of the high
proportion of nodes (73.8%) with canalyzing inputs (23). The
current network is not autonomous, so interpretation of K, P,
and the effects of canalyzing inputs on network behavior may be
different from the effects on the random, autonomous networks
in which these parameters have been studied (21, 23). However,
the fact that the network maps the wide ranging global inputs to
a relatively small number of global responses indicates that the
current system has a small number of attractors with large basins
of attraction, which is consistent with results with autonomous
networks with the similar connection parameters (21, 23).
The second prominent feature of the results is the biological
significance of both the global and individual outputs observed.
From the global output prospective, the output states 0000, 1000,
and 2000 are prominently represented because those outputs were
associated with 3,967 of the 10,000 random inputs. The outputs 1000
and 2000 represent quiescent states in which the outputs are
inactive, with the exception of Akt, a protein that must remain
active to suppress apoptosis (24). The state 0000 would be associ-
ated with apoptosis because Akt activity is very low. Looking at the
individual qualitative input–output relationships within these three
prominent global outputs, it can be seen that they are characterized
by low levels of extracellular matrix (ECM) and increasing levels of
EGF. This is consistent with the responsiveness of Akt activity to
EGF signaling found in the literature (25, 26) and consistent with
the input–output relationship of Akt and EGF presented in Fig. 2.
Despite the fact that EGF increases to high levels in the 2000
output, Erk activity does not increase. As expected from the known
dependence of EGF on ECM/Integrin stimulation (27), ECM levels
associated with these outputs is low, and Erk activity appears in the
Table 1. Outputs of the network and their average associated inputs
Global output
(ternary)
Count
Average input
Average output
ECM
EGF
ExtPump
qlig
ilig
slig
1213lig
IL1TNF
Stress
Akt
Erk
Cdc42
Rac
1000
2,346
26
26
54
49
48
50
49
2
2
19
3
2
1
2000
1,488
24
67
39
51
49
50
52
1
2
42
4
2
1
1011
952
79
21
55
49
51
50
52
2
2
19
4
19
16
2100
851
31
80
43
51
44
49
50
3
2
49
14
3
1
2110
833
58
78
45
50
53
50
48
2
2
47
15
17
5
2111
626
85
65
28
50
52
50
52
2
2
42
15
21
13
1010
463
50
34
70
46
57
50
44
2
2
21
4
15
5
2010
429
54
70
53
49
54
52
49
1
2
42
5
15
4
2121
361
89
71
55
49
57
47
42
2
2
42
17
37
14
1021
278
87
29
76
48
59
49
45
2
1
19
5
36
20
2011
232
80
46
29
51
55
50
55
1
2
35
6
19
14
1001
149
72
17
40
49
27
55
53
2
2
18
3
5
13
2120
136
69
85
74
48
56
50
38
2
2
52
18
37
6
0000
133
29
12
70
47
9
48
47
2
2
6
2
0
2
1111
112
82
34
48
53
48
54
51
4
2
24
11
21
16
1121
87
89
41
71
51
56
42
41
3
1
23
11
39
19
2021
78
87
61
63
46
63
57
49
1
1
38
6
36
14
1110
71
55
46
64
49
49
48
46
4
2
24
11
17
6
1100
58
35
50
57
51
39
52
46
4
2
25
10
4
2
2020
36
72
79
90
55
58
43
42
0
2
46
5
38
6
0011
30
80
13
90
53
16
52
47
1
2
7
2
18
18
1022
28
98
9
70
52
66
49
46
2
2
16
4
39
31
2200
26
26
97
77
47
26
52
45
4
2
68
37
1
0
2221
25
90
82
47
53
65
27
30
4
1
51
32
46
13
0001
25
79
14
73
54
2
54
45
2
3
6
2
4
14
2001
21
76
52
18
54
36
65
61
1
2
36
6
7
11
2101
20
82
73
22
53
28
40
51
3
2
45
13
6
11
2210
18
62
95
72
64
43
53
37
4
2
62
33
18
4
2220
17
74
95
75
50
53
52
41
4
2
61
33
43
6
1020
14
58
44
91
39
62
45
24
1
2
20
4
34
7
1012
13
98
5
49
46
62
37
51
1
2
18
3
26
30
2211
12
86
84
31
55
36
43
58
4
3
53
31
24
12
0010
10
48
8
90
62
17
47
52
2
2
7
2
12
6
1101
10
78
38
41
42
7
44
58
4
2
22
10
6
12
1120
4
61
56
85
55
50
34
5
4
1
26
14
40
6
0021
3
81
17
97
41
30
68
48
2
0
8
3
34
20
1122
3
96
6
47
53
85
74
21
3
1
21
13
48
31
0022
2
92
3
98
28
15
35
31
2
0
9
2
35
31
qlig, ilig, slig, and 1213lig are abbreviations for generic ligands for the respective G subunit of GPCR. Other abbreviations are as in the text. Standard
deviations were calculated but are not shown for clarity because the variance can be observed directly in the scatter plots in Fig. 3 and SI Fig. 5.
Helikar et al.
PNAS
February 12, 2008
vol. 105
no. 6
1915
CELL BIOLOGY
global outputs only when input levels of both ECM and EGF are
high. Similarly, global outputs with increased Cdc42 and Rac
activity correlate strongly with high levels of ECM; both Rac and
Cdc42 are classically associated with cytoskeletal regulation in
response to ECM (28). As a control, the network was randomly
‘‘rewired’’ 100 times, i.e., the inputs to each node were randomized
while preserving the in- and out-degrees as well as the logical table
of the individual nodes. The complementary control was also
performed 100 times, i.e., the graph of the network was held
constant while the logic was randomized. In both controls, the
number of outputs diminished to a trivial number of outputs (an
average effective number of 1.92 and 1.04, respectively), with no
biological significance in the correlation of input and output (see SI
Text). This indicates that both the graph and the logic are important
for the variety and biological significance of the outputs.
The facts that the individual qualitative input–output relation-
ships from Fig. 2 are present in Table 1 and that the global outputs
are biologically relevant support the validity of characterizing
ranges of output activity. However, the real power of this analysis
is the ability to observe how the system clusters combinations of
inputs and then maps them to the global outputs. To visualize this
mapping at a more detailed level, all 9,389 input vectors associated
with the 15 most frequent global outputs were subjected to principal
component analysis (PCA) (29). In the resulting plot, shown in Fig.
3A, each point (representing an individual input combination) is
colored according to the ternary output string with which it was
associated. It shows all 9,389 inputs together, and the different
colors appear to separate into discrete clusters. To confirm this, the
plots in Fig. 3B show several combinations of different colors to
indicate the degree of overlap of inputs associated with the most
common global outputs. The results show that when the random
input vectors are plotted in three-dimensional space based on their
values, they form a random scatter as expected. But when each
vector in that scatter plot is colored based on the global output with
which it is associated, all of the input vectors associated with a
particular output are not randomly scattered but, rather, clustered
in distinct areas with little overlap with inputs associated with other
outputs. Thus, this signal transduction system clusters neighbor-
hoods of input combinations into equivalence classes of global
outputs; i.e., all input combinations of the same color are consid-
ered to be functionally equivalent because they elicit the same
global output response. The 100 randomly rewired and random
logic control networks were also tested for separation and in
rewired networks where there was more than one output to test, the
number of outputs that demonstrated clustering of inputs was
greatly reduced and separation in the random logic networks was
eliminated. The details of PCA, how it was applied to the network,
statistical analysis, results with the rewired controls, and further
Fig. 3.
Scatter plots of all input
vectors associated with the first 15
global outputs of Table 1. (A) The
inputs associated with the 15 most
common outputs are plotted in
three dimensions by using principle
component analysis (PCA, see Ma-
terials and Methods). All 9,389 in-
puts plotted together, with each in-
put colored according to which of
the 15 outputs it is associated. It
appears that all inputs associated
with a given output (indicated by
the color) are clustered. (B) To verify
that the model uniquely clusters in-
puts based on associated outputs,
selected colored clusters in A are
plotted on separate axes so the sep-
aration of each cluster is visible. For
example, the 2,346 input values as-
sociated with the output 1000
(shown as black points) are clus-
tered with little overlap with input
values associated with outputs 2000
and 1011, as shown in the first plot.
Taken together, these results show
that the Boolean signal transduc-
tion model divides the input space
into distinct equivalence classes
that are associated with biologically
appropriate global outputs.
1916
www.pnas.orgcgidoi10.1073pnas.0705088105
Helikar et al.
discussion of the biological relevance of these results can be found
in Materials and Methods and SI Text.
The results presented show that this signal transduction network
model is capable of taking a wide array of random hormonal input
combinations and classify them into a relatively small number of
biologically appropriate, sharply defined equivalence classes of
global responses. This function can, by definition, be called pattern
recognition, a concept used in machine learning and neural net-
works (30, 31). The ability to recognize input patterns and classify
them is a decision-making function that is a form of information
processing. It involves dividing a multidimensional space into
associated classes, the boundaries of which must be carefully
determined to recognize inputs correctly based on their class
association (30). The practical effect of this type of processing is that
the very large number of combinations of possible hormonal inputs
to which a cell may be exposed (many of which are relatively
indistinct) are clustered by the signal transduction network accord-
ing to the much smaller number of global cellular responses that are
possible for a cell to make to each input. Thus, this network is able
to make decisions even in the face of less than clear-cut
environmental cues that are common in realistic environments.
To determine the robustness of signal transduction decision-
making, the above experiments were carried out with different
noise levels. For example, if in a given run, an input such as EGF
is set to 50% ON, no added noise would mean that the node is a
exactly 50% throughout the entire run. Adding 2% noise to a 50%
value would mean that the input varied chaotically between 48%
and 52% with an average of 50%. Five percent noise for that input
would result in the input varying chaotically between 45% and 55%,
and so on. In the above simulations, noise was added at what was
considered to be a normal background level of 2%. The results of
testing of other noise levels of up to 20% can be seen in SI Tables
7–9. It is clear that the pattern recognition ability of the network in
terms of global responses is nearly unaffected by even high levels of
noise. This surprising level of stability was verified by repeating the
individual input–output relationships of Fig. 2 with varying levels of
noise. Even at high levels of noise, the input–output relations
remained intact (data shown SI Fig. 6), confirming the ability of the
system to recognize patterns in even very noisy inputs.
It has long been noticed that complex, interconnected pathways
of biochemical signal transduction networks bear a resemblance to
parallel, distributed computer networks. This led to conjecture by
some that the overwhelming complexity of these networks might
not be an accident of evolution but, rather, a key characteristic of
a finely tuned information-processing system that is able to make
nontrivial decisions (5, 6, 31, 32). To test this hypothesis directly, we
have created a large-scale, literature-based, logically complete
model of a multifamily signal transduction network. The model was
then exposed to tens of thousands of different combinations of
environmental stimuli, and the global responses (i.e., combinations
of multiple outputs) were observed. The reason for this approach
was to look for emergent properties of the system by moving beyond
the exploration of the important and now well established dynamics
of specific, individual stimulus–response relationships (e.g., bist-
ability) and consider the higher-level relationships between multi-
ple stimuli and the corresponding global responses. This is the
essence of the systems approach.
The results clearly show that the network clustered the vast
majority of inputs into a small number of biologically appropriate
responses. This nonfuzzy partitioning of a space of random, noisy,
chaotic inputs into a small number of equivalence classes is a
hallmark of a pattern recognition machine and is strong evidence
that signal transduction networks are decision-making systems that
process information obtained at the membrane rather than simply
passing unmodified signals downstream.
Designing systems to perform sophisticated pattern recognition
is not a trivial task. Handwriting and face recognition are examples
of real-world, sophisticated pattern recognition where noisy inputs
must be correctly placed into a nonfuzzy, sharply defined equiva-
lence class (e.g., individual handwriting classified as a particular
character or an individual face recognized as an acquaintance); a
major goal of artificial intelligence research is to develop machines
that are capable of such tasks (30, 31). It should not be surprising
that cells would require a similar ability to perform sophisticated
pattern recognition. An individual cell is faced with any number of
stimuli in the form of chemical ligands binding to their cell-surface
receptors. These receptors are varied in type and number and form
a sensory system that enables a cell to sense and respond to its
environment. Given that any physical environment is chaotic, noisy,
and, at times, contradictory, it is clear that cells need the ability to
make decisions based on these types of inputs and that their survival
depends on that ability.
Finally, the results presented here use literature-based, Boolean
modeling of a large-scale biochemical system. All modeling meth-
ods have their downsides, and in the case of Boolean models it is
that the logic of each node must be expressed in terms of ON/OFF.
This seems counterintuitive to many biologists because it is known
that many signal transduction components do not have such simple
regulation. In reality, this is not a major obstacle to Boolean
modeling because proteins that exhibit more complex regulation
can be represented by multiple nodes, each representing a separate
activation state of the protein of interest (e.g., Raf in our network).
The only real downside to Boolean modeling of biochemical
systems is that, for any node with a large number of inputs (N), there
are 2N combinations of those inputs that must be accounted for in
each logic table. For most input combinations the ON/OFF state of
the protein can be derived from the literature in a straightforward
way. However, some combinations are not explicitly dealt with in
the literature and must be deduced indirectly. For this reason, we
do not consider the current network to be perfect. However, this
problem is not unsolvable; it only requires laboratory researchers to
test qualitatively the input combinations that are unknown. This can
be done exactly as the known combinations were determined, thus
requiring only an awareness for the need for this information rather
than entirely new laboratory methods. Additionally, our develop-
ment of tools that are able to input and retrieve continuous data to
and from the Boolean model means that the only aspect of the
model that is actually ON/OFF is the logic tables for each individual
node; once the logic is set, the model is used in the same way as
continuous models.
Continuous modeling, on the other hand, has the significant
problem of parameter estimation. Although there are also ways of
dealing with this problem, determination of the large number of
parameters of a large-scale network in vivo is a much more
complicated technical hurdle. All modeling methods also have
upsides, and the parameter-free nature of Boolean modeling is a
significant advantage, making it complementary to continuous
models used for exploring higher-order functions and emergent
properties of biological signal transduction networks.
Materials and Methods
The Boolean Model of Signal Transduction. To create a Boolean network, a set of
nodes must be identified and a logic table created for each node. The current
Boolean model of signal transduction was created by determining the complete
logic of the classical EGFR 3 Erk pathway. More detail on how the logic tables
were created for each node (as well as how it is possible to use Boolean modeling
for proteins that have more complex activation than simple ON/OFF) can be
found in SI Text. As guided by the literature, connections to other classical
pathways were included in the EGFR 3 Erk pathway until a relatively autono-
mous network of 130 nodes was created that included the RTK, GPCR, and
Integrin pathways. Given the highly interconnected nature of cytoplasmic pro-
tein networks, stopping at even 130 nodes meant that some interactions with
proteins outside these three families had to be ignored. However, these were
relatively minor compared with the interactions of the three incorporated path-
ways; these pathways are so intimately connected that they represent a func-
tioningsetofnodesthatwouldbeimpossibletoreducefurtherwithoutignoring
important interactions. Although the model is a nonspecific network in that it
Helikar et al.
PNAS
February 12, 2008
vol. 105
no. 6
1917
CELL BIOLOGY
does not represent any one specific cell type, nodes were not included in the
network unless they were generally expressed in a wide range of cell types.
However, once a node was included in the network, the best information on the
logic was used without regard to the cell type.
Inputs to the network are mostly the ligands of surface receptor nodes of
which there are seven; epidermal growth factor (EGF), the GPCR stimulators
(qlig, ilig, slig, i12/13lig), extracellular matrix (ECM), tumor necrosis fac-
tor/interleukin 1 (TNF/IL-1) an idealized hybrid receptor. In addition to those
ligands, ‘‘Stress’’ is an input representing environmental stress factors such as UV
light or reactive oxygen species (33, 34), and there is a nonregulated calcium
pump (external calcium pump). Calcium pumps are regulated mostly by calcium
and hormonal factors that are not included in the current network (35). The logic
for calcium regulation in the network inherently includes the calcium regulation
of the pump, but the other regulators cannot be accounted for. Therefore, the
calcium pump is considered to be an input and is set at multiple constant levels of
activity. Outputs of the network are nodes in the network whose outputs go out
to regulators of major cellular functions. These are (i) Akt, a major regulator of
apoptotic systems (24), (ii) the mitogen-activated protein kinase (MAPK) Erk, a
major regulator of cell division (34), and (iii) Rac and Cdc42, two important
regulatorsofcytoskeletalsystems(28).TheMAPK’sSAPKandp38arealsooutputs
of the network, but they respond to stress and TNF/IL-1 (33, 34), as documented
in SI Text. The experiments in the present work involve ‘‘stress-limited’’ inputs,
meaning that stress and TNF/IL-1 are at low, background levels. Other nodes can
be considered to be outputs of the network, and experiments with up to seven
different outputs were performed with very similar results.
Methods of Simulation. Although the logic of each node and input to the
networkisbinary,itispossibletointerpretintermediateactivitybylookingatthe
average ON value of each node when the system has reached a cycle, as all
Boolean models must do. Similarly, inputs can be set to a specific average ON by
putting the input on an appropriate cycle (for further details on this, see SI Text).
The actual simulations are performed by a Boolean simulation program de-
veloped by this group called ChemChains. ChemChains is a general Boolean
networksimulatorthatisabletoincorporateanynumberofnodesandtheirlogic
tables as well as any initial condition or input conditions and iterate the network
any desired number of times. The ChemChains program can be freely obtained
from J.A.R.
Adding Noise to the Inputs. Experiments are performed at different noise levels
by introducing a random noise component to the input that forces the input to
vary chaotically within a window around the set input level. The window sizes
vary from 2% to 20%, representing background noise to highly noisy inputs. In
these studies, all noise levels are tested and 2% noise is considered the standard
background levels. Noise was added to the inputs by adding or subtracting from
each input set point a percentage of the desired range. The percentage varied
randomlywithinthespecifiedrangebyusingarandomnumbergeneratorwithin
the ChemChains program.
Principal Component Analysis (PCA). PCA was done on all seven inputs and
projected onto three dimensions (accounting for 45% of variance of the system)
as shown in SI Fig. 5, where a more detailed explanation of PCA and the statistical
analysis of the results can be found. To capture more of the variance, various
numbers of inputs were tested and it was found that most of the pattern
recognition function could be observed by performing PCA on ECM, EGFR, and
the external calcium pump inputs and projecting onto three dimensions. This
accounts for 100% of the variance and makes the clustering of inputs the most
clearly visible. These results are shown in Fig. 3; however, they are not funda-
mentally different from the original seven-input PCA shown in SI Fig. 5.
ACKNOWLEDGMENTS. We thank J. Maloney for initial help with MAPLE pro-
gramming, J. Hamilton for creating the initial version of ChemChains, C. Ramey
for helpful discussions and creating the PCA program, and S. From for consulta-
tion on statistical methods. This work was supported by National Institutes of
Health Grant GM067272.
1. Jordan JD, Iyengar R (1998) Modes of interactions between signaling pathways.
Biochem Pharmacol 55:1347–1352.
2. Fambrough D, McClure K, Kazlauskas A, Lander ES (1999) Diverse signaling pathways
activated by growth factor receptors induce broadly overlapping, rather than inde-
pendent, sets of genes. Cell 97:727–741.
3. Jordan JD, Landau EM, Iyengar R (2000) Signaling networks: the origins of cellular
multitasking. Cell 103:193–200.
4. Bray D (1990) Intracellular signalling as a parallel distributed process. J Theor Biol
143:215–231.
5. Bray D (1995) Protein molecules as computational elements in living cells. Nature
376:307–312.
6. Schamel WW, Dick TP (1996) Signal transduction: Specificity of growth factors ex-
plained by parallel distributed processing. Med Hypotheses 47:249–255.
7. Bhalla US, Iyengar R (1999) Emergent properties of networks of biological signaling
pathways. Science 283:381–387.
8. Fernandez P, Sole RV (2005) in Power Laws. Scale-Free Networks and Genome Biology,
eds Koonin EV, Wolf YI, Karev GP (Springer, New York), pp 206–224.
9. Hopfield JJ (1994) Physics, computation, and why biology looks so different. J Theor
Biol 171:53–60.
10. Hartwell LH, Hopfield JJ, Leibler S, Murray AW (1999) From molecular to modular cell
biology. Nature 402:C47–52.
11. Kitano H (2002) Systems biology: A brief overview. Science 295:1662–1664.
12. Cho K, Wolkenhauer O (2003) Analysis and modelling of signal transduction pathways
in systems biology. Biochem Soc Trans 31:1503–1509.
13. Van Regenmortel MHV (2004) Reductionism and complexity in molecular biology.
Scientists now have the tools to unravel biological and overcome the limitations of
reductionism. EMBO Rep 5:1016–1020.
14. Albert R (2005) Scale-free networks in cell biology. J Cell Sci 118:4947–4957.
15. Zhu H, Huang S, Dhar P (2003) The next step in systems biology: Simulating the
temporospatial dynamics of molecular network. BioEssays 26:68–72.
16. Rodriguez-Fernandez M, Mendes P, Banga JR (2006) A hybrid approach for efficient
and robust parameter estimation in biochemical pathways. Biosystems 83:248–265.
17. Chou I, Martens H, Voit EO (2006) Parameter estimation in biochemical systems models
with alternating regression. Theor Biol Med Model 3:25.
18. Koh G, Teong HFC, Cle´ment M, Hsu D, Thiagarajan PS (2006) A decompositional
approach to parameter estimation in pathway modeling: A case study of the Akt and
MAPK pathways and their crosstalk. Bioinformatics 22:e271–80.
19. Huang S (2001) Genomics, complexity and drug discovery: Insights from Boolean
network models of cellular regulation. Pharmacogenomics 2:203–222.
20. Shmulevich I, Dougherty ER, Zhang W (2002) From Boolean to probabilistic Boolean
networks as models of genetic regulatory networks. Proc IEEE 90:1778–1792.
21. Kauffman SA (1993) in The Origins of Order (Oxford Univ Press, New York), pp 188–235.
22. Aldana M, Cluzel P (2003) A natural class of robust networks. Proc Natl Acad Sci USA
100:8710–8714.
23. Kauffman SA (1993) in The Origins of Order (Oxford Univ Press, New York), pp
441–481.
24. Kumar CC, Madison V (2005) AKT crystal structure and AKT-specific inhibitors. Onco-
gene 24:7493–7501.
25. Du K, Tsichlis PN (2005) Regulation of the Akt kinase by interacting proteins. Oncogene
24:7401–7409.
26. Kassenbrock CK, Hunter S, Garl P, Johnson GL, Anderson SM (2002) Inhibition of Src
family kinases blocks epidermal growth factor (EGF)-induced activation of Akt, phos-
phorylation of c-Cbl, and ubiquitination of the EGF receptor. J Biol Chem 277:24967–
24975.
27. Edin ML, Juliano RL (2005) Raf-1 serine 338 phosphorylation plays a key role in
adhesion-dependent activation of extracellular signal-regulated kinase by epidermal
growth factor. Mol Cell Biol 25:4466–4475.
28. Price LS, Leng J, Schwartz MA, Bokoch GM (1998) Activation of Rac and Cdc42 by
integrins mediates cell spreading. Mol Biol Cell 9:1863–1871.
29. Wall ME, Rechtsteiner A, Rocha LM (2003) in A practical approach to microarray data
analysis, eds Berrar DP, Dubitzky W, Granzow M (Kluwer, Boston), pp 91–109.
30. Haykin S (1999) in Neural Networks, A Comprehensive Foundation (Prentice–Hall,
Englewood Cliffs, NJ), pp 66–67.
31. Bray D (2003) Molecular networks: The top-down view. Science 301:1864–1865.
32. Ma’ayan A, et al. (2005) Formation of regulatory patterns during signal propagation
in a mammalian cellular network. Science 309:1078–1083.
33. Takeda K, Matsuzawa A, Nishitoh H, Ichijo H (2003) Roles of MAPKKK ASK1 in
stress-induced cell death. Cell Struct Funct 28:23–29.
34. Roux PP, Blenis J (2004) ERK and p38 MAPK-activated protein kinases: A family of
protein kinases with diverse biological functions. Microbiol Mol Biol Rev 68:320–344.
35. Strehler EE, Treiman M (2004) Calcium pumps of plasma membrane and cell interior.
Curr Mol Med 4:323–335.
36. Brakebusch C, Bouvard D, Stanchi F, Sakai T, Fa¨ssler R (2002) Integrins in invasive
growth. J Clin Invest 109:999–1006.
37. Milligan G, White JH (2001) Protein–protein interactions at G-protein-coupled recep-
tors. Trends Pharmacol Sci 22:513–518.
38. Selbie LA, Hill SJ (1998) G protein-coupled-receptor cross-talk: the fine-tuning of
multiple receptor-signalling pathways. Trends Pharmacol Sci 19:87–93.
39. Naor Z, Benard O, Seger R (2000) Activation of MAPK cascades by G-protein-coupled
receptors: the case of gonadotropin-releasing hormone receptor. Trends Endocrinol
Metab 11:91–99.
40. Gschwind A, Zwick E, Prenzel N, Leserer M, Ullrich A (2001) Cell communication
networks: epidermal growth factor receptor transactivation as the paradigm for
interreceptor signal transmission. Oncogene 20:1594–1600.
41. Sridhar SS, Hedley D, Siu LL (2005) Raf kinase as a target for anticancer therapeutics.
Mol Cancer Ther 4:677–685.
1918
www.pnas.orgcgidoi10.1073pnas.0705088105
Helikar et al.
|
18250321
|
PTP1b = ( NOT ( ( EGFR AND ( ( ( EGF ) ) ) ) OR ( Stress ) ) ) OR NOT ( EGFR OR EGF OR Stress )
Csk = ( Cbp AND ( ( ( Gbg_i OR PKA OR Gbg_1213 OR Gbg_q ) ) OR ( ( NOT Gbg_i AND NOT PKA AND NOT SHP2 AND NOT Gbg_1213 AND NOT Gbg_q ) ) ) ) OR ( ( Fak AND ( ( ( Src AND Cbp ) ) ) ) AND NOT ( SHP2 ) )
PI3K = ( Gbg_i ) OR ( Gab1 ) OR ( Fak ) OR ( Ras ) OR ( Crk ) OR ( EGFR AND ( ( ( Src ) ) ) )
PIP_4 = ( ( ( PTEN AND ( ( ( NOT PIP_4 ) ) AND ( ( PIP2_34 ) ) ) ) AND NOT ( PI5K AND ( ( ( PIP_4 ) ) ) ) ) AND NOT ( PI3K AND ( ( ( PIP_4 ) ) ) ) ) OR ( ( ( PIP_4 AND ( ( ( NOT PI3K ) ) AND ( ( NOT PI5K ) ) ) ) AND NOT ( PI5K AND ( ( ( PIP_4 ) ) ) ) ) AND NOT ( PI3K AND ( ( ( PIP_4 ) ) ) ) ) OR ( ( ( PI4K AND ( ( ( NOT PIP_4 ) ) ) ) AND NOT ( PI5K AND ( ( ( PIP_4 ) ) ) ) ) AND NOT ( PI3K AND ( ( ( PIP_4 ) ) ) ) )
Talin = ( PIP2_45 AND ( ( ( NOT Talin ) ) ) ) OR ( Talin AND ( ( ( NOT Src ) ) ) )
Gbg_s = ( Gas ) OR ( alpha_sR AND ( ( ( NOT Gbg_s ) ) AND ( ( NOT Gas ) ) ) )
TAO_12 = ( Stress )
Mekk3 = ( ( Trafs ) AND NOT ( Gab1 ) ) OR ( ( IL1_TNFR ) AND NOT ( Gab1 ) ) OR ( ( Rac ) AND NOT ( Gab1 ) )
MKPs = ( p38 AND ( ( ( cAMP ) ) ) ) OR ( SAPK AND ( ( ( cAMP ) ) ) ) OR ( Erk AND ( ( ( cAMP ) ) ) )
Tab_12 = ( ( Trafs ) AND NOT ( p38 ) )
Gas = ( alpha_sR AND ( ( ( NOT Gas ) ) AND ( ( NOT PKA ) ) AND ( ( NOT Gbg_s ) ) ) ) OR ( Gbg_s AND ( ( ( Gas ) ) AND ( ( NOT RGS ) ) ) )
p38 = ( ( ( MKK6 ) AND NOT ( PP2A ) ) AND NOT ( MKPs ) ) OR ( ( ( MKK3 ) AND NOT ( PP2A ) ) AND NOT ( MKPs ) ) OR ( ( ( Sek1 ) AND NOT ( PP2A ) ) AND NOT ( MKPs ) )
Rho = ( Rho AND ( ( ( NOT PKA AND NOT p190RhoGAP AND NOT Graf ) ) ) ) OR ( p115RhoGEF AND ( ( ( NOT Rho AND NOT RhoGDI ) ) ) )
PP2A = ( NOT ( ( EGFR ) ) ) OR NOT ( EGFR )
Gab1 = ( ( Gab1 AND ( ( ( EGFR AND PIP3_345 ) ) ) ) AND NOT ( SHP2 ) ) OR ( ( Grb2 AND ( ( ( EGFR ) AND ( ( ( NOT Gab1 ) ) ) ) ) ) AND NOT ( SHP2 ) )
PIP2_34 = ( PIP2_34 AND ( ( ( NOT PTEN ) ) AND ( ( NOT PI5K ) ) ) ) OR ( PI4K AND ( ( ( NOT PIP2_34 ) ) AND ( ( PI3K ) ) ) )
PIP3_345 = ( ( PI3K AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( PTEN AND ( ( ( PIP3_345 ) ) ) ) ) OR ( ( PI5K AND ( ( ( PIP2_34 ) ) ) ) AND NOT ( PTEN AND ( ( ( PIP3_345 ) ) ) ) )
Vinc = ( Actin AND ( ( ( NOT PIP2_45 ) ) AND ( ( Vinc AND Talin ) ) ) ) OR ( Talin AND ( ( ( Src ) ) ) )
IP3R1 = ( ( ( ( Gbg_i ) AND NOT ( CaM AND ( ( ( Ca ) ) AND ( ( IP3R1 ) ) ) ) ) AND NOT ( IP3R1 AND ( ( ( Gbg_i AND CaM ) ) AND ( ( NOT PP2A AND NOT PKA AND NOT IP3 AND NOT Ca ) ) ) ) ) AND NOT ( Ca AND ( ( ( NOT IP3 ) ) AND ( ( IP3R1 ) ) ) ) ) OR ( ( ( IP3 AND ( ( ( Ca ) ) ) ) AND NOT ( CaM AND ( ( ( Ca ) ) AND ( ( IP3R1 ) ) ) ) ) AND NOT ( Ca AND ( ( ( NOT IP3 ) ) AND ( ( IP3R1 ) ) ) ) ) OR ( ( ( ( PKA ) AND NOT ( CaM AND ( ( ( Ca ) ) AND ( ( IP3R1 ) ) ) ) ) AND NOT ( Ca AND ( ( ( NOT IP3 ) ) AND ( ( IP3R1 ) ) ) ) ) AND NOT ( PP2A AND ( ( ( IP3R1 ) ) ) ) )
PTEN = ( ( Stress ) AND NOT ( Src AND ( ( ( PTEN ) ) ) ) ) OR ( ( Pix_Cool AND ( ( ( Cdc42 ) ) AND ( ( PI3K ) ) AND ( ( Rho ) ) ) ) AND NOT ( Src AND ( ( ( PTEN ) ) ) ) )
ASK1 = ( Trx )
AA = ( PLA2 )
PKC = ( ( AA AND ( ( ( Ca ) ) AND ( ( PKC_primed ) ) ) ) AND NOT ( Trx AND ( ( ( PKC ) ) ) ) ) OR ( ( PKC AND ( ( ( NOT PP2A ) ) AND ( ( NOT Trx ) ) ) ) AND NOT ( Trx AND ( ( ( PKC ) ) ) ) ) OR ( ( DAG AND ( ( ( Ca ) ) AND ( ( PKC_primed ) ) ) ) AND NOT ( Trx AND ( ( ( PKC ) ) ) ) )
Arp_23 = ( WASP )
Gai = ( Gbg_i AND ( ( ( Gai ) ) AND ( ( NOT RGS ) ) ) ) OR ( alpha_iR AND ( ( ( NOT Gbg_i AND NOT Gai ) ) ) ) OR ( PKA AND ( ( ( NOT Gai ) ) AND ( ( NOT Gbg_i ) ) AND ( ( NOT alpha_sR ) ) AND ( ( alpha_sL ) ) ) )
Cas = ( ( Src AND ( ( ( Fak ) ) ) ) AND NOT ( PTPPEST AND ( ( ( Cas ) ) ) ) )
ILK = ( PIP3_345 )
IP3 = ( PLC_B AND ( ( ( PIP2_45 ) ) ) ) OR ( PLC_g AND ( ( ( PIP2_45 ) ) ) )
PLA2 = ( PIP3_345 AND ( ( ( PIP2_45 ) ) AND ( ( CaMK ) ) ) ) OR ( PIP2_45 AND ( ( ( Erk ) ) AND ( ( PIP3_345 ) ) ) ) OR ( CaMK AND ( ( ( Ca ) ) ) ) OR ( Erk AND ( ( ( Ca ) ) ) )
alpha_1213R = ( ( alpha_1213L ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_1213R AND NOT alpha_1213L AND NOT alpha_1213R ) ) OR ( ( Palpha_1213R ) ) ) ) ) OR ( ( Palpha_1213R AND ( ( ( NOT B_Arrestin ) ) ) ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_1213R AND NOT alpha_1213L AND NOT alpha_1213R ) ) OR ( ( Palpha_1213R ) ) ) ) ) OR ( ( alpha_1213R ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_1213R AND NOT alpha_1213L AND NOT alpha_1213R ) ) OR ( ( Palpha_1213R ) ) ) ) )
PI5K = ( PA ) OR ( PI5K AND ( ( ( Talin ) ) ) ) OR ( RhoK ) OR ( Src AND ( ( ( Fak ) ) AND ( ( NOT PI5K ) ) AND ( ( NOT Talin ) ) ) ) OR ( ARF )
Sos = ( ( Grb2 AND ( ( ( PIP3_345 ) ) ) ) AND NOT ( Erk ) ) OR ( Nck AND ( ( ( Crk ) ) AND ( ( PIP3_345 ) ) ) )
PIP2_45 = ( PTEN AND ( ( ( PIP3_345 ) ) ) ) OR ( PI4K AND ( ( ( PI5K ) ) ) ) OR ( PIP2_45 )
Trx = ( Stress ) OR ( Trafs )
GRK = ( ( ( Gbg_i AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) ) OR ( ( ( Gbg_q AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) ) OR ( ( ( B_Arrestin AND ( ( ( Src ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) ) OR ( ( ( Gbg_1213 AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) ) OR ( ( ( Gbg_s AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) )
p190RhoGAP = ( Src AND ( ( ( NOT p190RhoGAP ) ) OR ( ( Fak ) ) OR ( ( NOT p120RasGAP ) ) ) ) OR ( Fak AND ( ( ( Src ) ) ) )
Myosin = ( ( ILK AND ( ( ( NOT MLCP ) ) OR ( ( NOT Myosin ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( MLCK AND ( ( ( NOT MLCP ) ) AND ( ( CaM ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( PAK AND ( ( ( NOT MLCP ) ) OR ( ( NOT Myosin ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( RhoK AND ( ( ( NOT MLCP ) ) OR ( ( NOT Myosin ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( CaM AND ( ( ( NOT Myosin ) ) AND ( ( MLCK ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( Myosin AND ( ( ( NOT MLCP ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) )
alpha_qR = ( ( alpha_qL ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_iR AND NOT alpha_qL AND NOT alpha_qR ) ) OR ( ( Palpha_iR ) ) ) ) ) OR ( ( Palpha_iR AND ( ( ( NOT B_Arrestin ) ) ) ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_iR AND NOT alpha_qL AND NOT alpha_qR ) ) OR ( ( Palpha_iR ) ) ) ) ) OR ( ( alpha_qR ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_iR AND NOT alpha_qL AND NOT alpha_qR ) ) OR ( ( Palpha_iR ) ) ) ) )
IL1_TNFR = ( IL1_TNF )
Tiam = ( Src AND ( ( ( Rap1 OR Ras OR PIP2_45 ) ) AND ( ( PIP3_345 OR PIP2_34 ) ) ) ) OR ( PKC AND ( ( ( PIP3_345 OR PIP2_34 ) ) AND ( ( Rap1 OR Ras OR PIP2_45 ) ) ) ) OR ( CaMK AND ( ( ( Rap1 OR Ras OR PIP2_45 ) ) AND ( ( PIP3_345 OR PIP2_34 ) ) ) )
NIK = ( TAK1 ) OR ( Nck )
Cdc42 = ( ( Cdc42 AND ( ( ( Pix_Cool ) ) AND ( ( NOT RhoGDI ) ) ) ) AND NOT ( RhoGDI AND ( ( ( Src ) ) ) ) ) OR ( ( Pix_Cool AND ( ( ( NOT Cdc42 AND NOT Rac ) ) AND ( ( PAK AND Gbg_i ) ) ) ) AND NOT ( RhoGDI AND ( ( ( Src ) ) ) ) )
PAK = ( ( ( Src AND ( ( ( PAK ) AND ( ( ( Cdc42 OR Rac ) ) ) ) ) ) AND NOT ( PKA ) ) AND NOT ( PTP1b ) ) OR ( ( Rac AND ( ( ( Grb2 ) ) OR ( ( Nck ) AND ( ( ( NOT Akt ) ) ) ) ) ) AND NOT ( PKA ) ) OR ( ( Cdc42 AND ( ( ( Nck ) AND ( ( ( NOT Akt ) ) ) ) OR ( ( Grb2 ) ) ) ) AND NOT ( PKA ) )
Rap1 = ( CaMK AND ( ( ( NOT Gai OR NOT Rap1 ) ) AND ( ( Src AND cAMP ) ) ) ) OR ( PKA AND ( ( ( NOT Gai OR NOT Rap1 ) ) AND ( ( Src AND cAMP ) ) ) )
p120RasGAP = ( ( ( PIP3_345 ) AND NOT ( Src ) ) AND NOT ( Fak ) ) OR ( ( ( Ca ) AND NOT ( Src ) ) AND NOT ( Fak ) ) OR ( ( ( PIP2_34 ) AND NOT ( Src ) ) AND NOT ( Fak ) ) OR ( ( ( PIP2_45 ) AND NOT ( Src ) ) AND NOT ( Fak ) ) OR ( ( ( ( EGFR ) AND NOT ( SHP2 ) ) AND NOT ( Src ) ) AND NOT ( Fak ) )
p115RhoGEF = ( Ga_1213 AND ( ( ( PIP3_345 ) ) ) )
Ral = ( CaM ) OR ( RalGDS ) OR ( AND_34 )
Graf = ( Fak AND ( ( ( Src ) ) ) )
GCK = ( Trafs )
alpha_sR = ( ( alpha_sR ) AND NOT ( B_Arrestin AND ( ( ( Palpha_sR ) ) OR ( ( NOT alpha_sR AND NOT Palpha_sR AND NOT alpha_sL ) ) ) ) ) OR ( ( alpha_sL ) AND NOT ( B_Arrestin AND ( ( ( Palpha_sR ) ) OR ( ( NOT alpha_sR AND NOT Palpha_sR AND NOT alpha_sL ) ) ) ) ) OR ( ( Palpha_sR AND ( ( ( NOT B_Arrestin ) ) ) ) AND NOT ( B_Arrestin AND ( ( ( Palpha_sR ) ) OR ( ( NOT alpha_sR AND NOT Palpha_sR AND NOT alpha_sL ) ) ) ) )
PI4K = ( Rho ) OR ( PKC ) OR ( ARF ) OR ( Gai ) OR ( Gaq )
Gbg_q = ( Gaq ) OR ( alpha_qR AND ( ( ( NOT Gaq ) ) AND ( ( NOT Gbg_q ) ) ) )
Ca = ( ( IP3R1 ) AND NOT ( ExtPump ) )
Trafs = ( IL1_TNFR )
Raf_Loc = ( Raf_Loc AND ( ( ( NOT Raf ) ) ) ) OR ( Ras AND ( ( ( Raf_DeP ) ) AND ( ( NOT Raf_Loc ) ) ) )
SHP2 = ( Gab1 )
Actin = ( Arp_23 AND ( ( ( Myosin ) ) ) )
Gaq = ( Gaq AND ( ( ( Gbg_q ) ) AND ( ( NOT RGS AND NOT PLC_B ) ) ) ) OR ( alpha_qR AND ( ( ( NOT Gaq ) AND ( ( ( NOT Gbg_q ) ) ) ) ) )
alpha_iR = ( ( alpha_iL ) AND NOT ( B_Arrestin AND ( ( ( Palpha_iR ) ) OR ( ( NOT alpha_iL AND NOT Palpha_iR AND NOT alpha_iR ) ) ) ) ) OR ( ( Palpha_iR AND ( ( ( NOT B_Arrestin ) ) ) ) AND NOT ( B_Arrestin AND ( ( ( Palpha_iR ) ) OR ( ( NOT alpha_iL AND NOT Palpha_iR AND NOT alpha_iR ) ) ) ) ) OR ( ( alpha_iR ) AND NOT ( B_Arrestin AND ( ( ( Palpha_iR ) ) OR ( ( NOT alpha_iL AND NOT Palpha_iR AND NOT alpha_iR ) ) ) ) )
Mekk4 = ( Cdc42 ) OR ( Rac )
B_Parvin = ( ILK )
MKK6 = ( Mekk4 AND ( ( ( ASK1 ) ) ) ) OR ( MLK3 AND ( ( ( ASK1 ) ) ) ) OR ( PAK AND ( ( ( ASK1 ) ) ) ) OR ( TAK1 AND ( ( ( ASK1 ) ) ) ) OR ( Tpl2 AND ( ( ( ASK1 ) ) ) ) OR ( TAO_12 AND ( ( ( ASK1 ) ) ) )
Palpha_iR = ( alpha_iR AND ( ( ( GRK ) ) ) )
PLD = ( Rho AND ( ( ( Actin ) AND ( ( ( PIP2_45 ) ) OR ( ( PIP3_345 ) ) ) ) AND ( ( NOT ARF ) ) ) ) OR ( PKC AND ( ( ( Actin ) AND ( ( ( PIP3_345 ) ) OR ( ( PIP2_45 ) ) ) ) AND ( ( NOT ARF ) ) ) ) OR ( ARF AND ( ( ( PIP2_45 ) ) OR ( ( PIP3_345 ) ) ) ) OR ( Rac AND ( ( ( NOT ARF ) ) AND ( ( Actin ) AND ( ( ( PIP2_45 ) ) OR ( ( PIP3_345 ) ) ) ) ) ) OR ( Cdc42 AND ( ( ( Actin ) AND ( ( ( PIP2_45 ) ) OR ( ( PIP3_345 ) ) ) ) AND ( ( NOT ARF ) ) ) )
MLCP = ( ( ( ( ( ( PKA AND ( ( ( RhoK ) ) ) ) AND NOT ( ILK ) ) AND NOT ( Raf ) ) AND NOT ( PAK ) ) AND NOT ( PKC ) ) ) OR NOT ( PAK OR PKA OR Raf OR RhoK OR ILK OR PKC )
DGK = ( PKC AND ( ( ( DAG ) ) ) ) OR ( Src AND ( ( ( Ca AND PA ) ) ) ) OR ( EGFR )
cAMP = ( ( AC ) AND NOT ( PDE4 ) ) OR ( ( cAMP ) AND NOT ( PDE4 ) )
RasGRF_GRP = ( CaM AND ( ( ( Cdc42 ) ) ) ) OR ( DAG AND ( ( ( Cdc42 ) ) ) )
TAK1 = ( Tab_12 )
Grb2 = ( Src AND ( ( ( Fak ) ) ) ) OR ( EGFR ) OR ( Shc )
Sek1 = ( Mekk4 AND ( ( ( ASK1 ) ) ) ) OR ( MLK1 AND ( ( ( ASK1 ) ) ) ) OR ( MLK2 AND ( ( ( ASK1 ) ) ) ) OR ( MLK3 AND ( ( ( ASK1 ) ) ) ) OR ( TAK1 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk1 AND ( ( ( ASK1 ) ) ) ) OR ( Tpl2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk3 AND ( ( ( ASK1 ) ) ) )
ARF = ( PIP2_45 ) OR ( PIP3_345 )
MLK3 = ( Rac ) OR ( IL1_TNFR ) OR ( Cdc42 )
Cbp = ( ( Src ) AND NOT ( SHP2 ) )
PKA = ( ( PDK1 AND ( ( ( cAMP ) ) ) ) AND NOT ( PP2A AND ( ( ( PKA ) ) ) ) ) OR ( ( PKA AND ( ( ( cAMP ) ) ) ) AND NOT ( PP2A AND ( ( ( PKA ) ) ) ) )
AC = ( Integrins AND ( ( ( ECM ) AND ( ( ( Gas ) ) AND ( ( Gbg_i ) ) ) ) ) )
WASP = ( ( Fak AND ( ( ( Grb2 OR Nck OR PIP2_45 ) ) AND ( ( Cdc42 AND Crk ) ) ) ) AND NOT ( PTPPEST ) ) OR ( ( Src AND ( ( ( Grb2 OR Nck OR PIP2_45 ) ) AND ( ( Cdc42 AND Crk ) ) ) ) AND NOT ( PTPPEST ) ) OR ( ( Cdc42 AND ( ( ( NOT PTPPEST AND NOT Crk ) ) AND ( ( Fak OR Src ) ) AND ( ( Grb2 OR Nck OR PIP2_45 ) ) ) ) AND NOT ( PTPPEST ) )
Rac = ( ( ( ( RasGRF_GRP AND ( ( ( ECM AND Integrins ) ) ) ) AND NOT ( RhoGDI AND ( ( ( NOT PAK ) ) ) ) ) AND NOT ( p190RhoGAP AND ( ( ( Rac ) ) ) ) ) AND NOT ( RalBP1 AND ( ( ( Rac ) ) ) ) ) OR ( ( Pix_Cool AND ( ( ( PAK AND Gbg_i ) AND ( ( ( NOT Cdc42 AND NOT Rac ) ) AND ( ( ECM AND Integrins ) ) ) ) OR ( ( NOT Gbg_i ) AND ( ( ( Cdc42 ) ) AND ( ( NOT Rac ) ) AND ( ( ECM AND Integrins ) ) ) ) OR ( ( NOT PAK ) AND ( ( ( NOT RasGRF_GRP AND NOT DOCK180 AND NOT Tiam ) ) AND ( ( Cdc42 ) ) AND ( ( NOT RhoGDI ) ) AND ( ( ECM AND Integrins ) ) AND ( ( NOT Rac ) ) ) ) ) ) AND NOT ( RhoGDI AND ( ( ( NOT PAK ) ) ) ) ) OR ( ( ( ( Tiam AND ( ( ( ECM AND Integrins ) ) ) ) AND NOT ( RhoGDI AND ( ( ( NOT PAK ) ) ) ) ) AND NOT ( p190RhoGAP AND ( ( ( Rac ) ) ) ) ) AND NOT ( RalBP1 AND ( ( ( Rac ) ) ) ) ) OR ( ( ( ( DOCK180 AND ( ( ( ECM AND Integrins ) ) ) ) AND NOT ( RhoGDI AND ( ( ( NOT PAK ) ) ) ) ) AND NOT ( p190RhoGAP AND ( ( ( Rac ) ) ) ) ) AND NOT ( RalBP1 AND ( ( ( Rac ) ) ) ) )
Gbg_1213 = ( Ga_1213 ) OR ( alpha_1213R AND ( ( ( NOT Ga_1213 ) ) AND ( ( NOT Gbg_1213 ) ) ) )
PDK1 = ( p90RSK ) OR ( Src )
Raf = ( Ras AND ( ( ( Raf ) ) ) ) OR ( Src AND ( ( ( NOT Raf ) ) AND ( ( PAK AND Raf_Loc AND RKIP ) ) ) ) OR ( Raf AND ( ( ( NOT PKA AND NOT Erk AND NOT Akt ) ) ) ) OR ( PAK AND ( ( ( NOT Erk AND NOT Akt AND NOT Ras ) ) AND ( ( Raf ) ) ) )
Akt = ( CaMKK AND ( ( ( NOT Akt ) ) AND ( ( Src AND ILK ) ) AND ( ( PIP3_345 OR PIP2_34 ) ) ) ) OR ( Akt AND ( ( ( NOT PP2A ) ) ) ) OR ( PDK1 AND ( ( ( NOT Akt ) ) AND ( ( PIP3_345 OR PIP2_34 ) ) AND ( ( Src AND ILK ) ) ) )
AND_34 = ( Cas )
RhoK = ( Rho )
PA = ( PLD )
PTPPEST = ( ( ( Integrins AND ( ( ( ECM ) ) ) ) AND NOT ( PKA ) ) AND NOT ( PKC ) )
SAPK = ( ( ( MKK7 ) AND NOT ( MKPs AND ( ( ( SAPK ) ) ) ) ) AND NOT ( PP2A AND ( ( ( SAPK ) ) ) ) ) OR ( ( ( Sek1 ) AND NOT ( MKPs AND ( ( ( SAPK ) ) ) ) ) AND NOT ( PP2A AND ( ( ( SAPK ) ) ) ) )
Palpha_sR = ( alpha_sR AND ( ( ( GRK ) ) ) )
B_Arrestin = ( Palpha_iR ) OR ( Palpha_qR ) OR ( Palpha_1213R ) OR ( Palpha_sR )
Tpl2 = ( Trafs )
Ga_1213 = ( Ga_1213 AND ( ( ( NOT p115RhoGEF ) ) AND ( ( Gbg_1213 ) ) ) ) OR ( alpha_1213R AND ( ( ( NOT Ga_1213 AND NOT Gbg_1213 ) ) ) )
Pix_Cool = ( PIP3_345 AND ( ( ( B_Parvin ) ) ) ) OR ( PIP2_34 AND ( ( ( B_Parvin ) ) ) )
Integrins = ( Talin AND ( ( ( NOT Integrins AND NOT ILK ) ) AND ( ( ECM ) ) ) ) OR ( Src AND ( ( ( NOT PP2A AND NOT ECM AND NOT Talin AND NOT Integrins AND NOT ILK ) ) ) ) OR ( PP2A AND ( ( ( NOT Integrins ) ) AND ( ( ECM AND Talin AND ILK ) ) ) ) OR ( Integrins AND ( ( ( NOT Src AND NOT ILK ) ) ) )
CaM = ( Ca )
Fak = ( ( Integrins AND ( ( ( Talin ) ) ) ) AND NOT ( PTEN AND ( ( ( Fak ) ) ) ) ) OR ( ( Src AND ( ( ( Fak ) ) ) ) AND NOT ( PTEN AND ( ( ( Fak ) ) ) ) )
Shc = ( ( EGFR AND ( ( ( Fak AND Src ) ) ) ) AND NOT ( Shc AND ( ( ( EGFR AND Fak AND Src AND PTEN ) ) ) ) )
Mekk2 = ( PI3K AND ( ( ( EGFR ) ) AND ( ( NOT Mekk2 ) ) ) ) OR ( PLC_g AND ( ( ( NOT Mekk2 ) ) AND ( ( EGFR ) ) ) ) OR ( Src AND ( ( ( EGFR ) ) AND ( ( NOT Mekk2 ) ) ) ) OR ( Grb2 AND ( ( ( EGFR ) ) AND ( ( NOT Mekk2 ) ) ) )
PTPa = ( PKC )
PLC_g = ( Fak AND ( ( ( Src ) ) AND ( ( NOT EGFR AND NOT PIP3_345 AND NOT PA AND NOT AA ) ) ) ) OR ( Src AND ( ( ( PIP3_345 AND Fak ) ) ) ) OR ( ( EGFR AND ( ( ( PIP3_345 ) ) ) ) AND NOT ( PA AND ( ( ( NOT Fak AND NOT Src ) ) AND ( ( AA ) ) ) ) )
Raf_DeP = ( PP2A AND ( ( ( NOT Raf_DeP ) ) AND ( ( Raf_Rest ) ) ) ) OR ( Raf_DeP AND ( ( ( NOT Raf_Loc ) ) ) )
p90RSK = ( Erk AND ( ( ( PDK1 ) ) AND ( ( NOT p90RSK ) ) ) )
PKC_primed = ( PKC AND ( ( ( PDK1 ) ) AND ( ( NOT PKC_primed ) ) ) ) OR ( PKC_primed AND ( ( ( NOT PKC ) ) ) ) OR ( PDK1 AND ( ( ( NOT PKC ) ) ) )
PLC_B = ( ( Gbg_i AND ( ( ( PLC_B ) ) ) ) AND NOT ( PKA AND ( ( ( NOT Gaq ) ) ) ) ) OR ( Gaq )
Raf_Rest = ( ( Raf_Rest AND ( ( ( NOT Raf_DeP ) ) ) ) OR ( Raf_DeP AND ( ( ( NOT Raf AND NOT Raf_Rest ) ) ) ) ) OR NOT ( Raf_Rest OR Raf OR Raf_DeP )
MLCK = ( ( ( CaM AND ( ( ( NOT PAK ) ) AND ( ( NOT PKA ) ) ) ) AND NOT ( PAK ) ) AND NOT ( PKA ) ) OR ( ( ( Erk AND ( ( ( NOT PAK ) ) AND ( ( NOT PKA ) ) ) ) AND NOT ( PAK ) ) AND NOT ( PKA ) )
Crk = ( ( Cas AND ( ( ( Fak OR Src ) ) ) ) AND NOT ( PTPPEST ) )
Palpha_1213R = ( alpha_1213R AND ( ( ( GRK ) ) ) )
Mek = ( ( PAK AND ( ( ( Tpl2 ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Tpl2 ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Mekk1 AND ( ( ( Raf ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Raf AND ( ( ( Tpl2 ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Mekk2 AND ( ( ( Raf ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Mekk3 AND ( ( ( Raf ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) )
PDE4 = ( B_Arrestin AND ( ( ( NOT Erk ) ) ) ) OR ( PKA AND ( ( ( B_Arrestin ) ) ) )
CaMK = ( CaMKK AND ( ( ( CaM ) ) ) )
Mekk1 = ( Rho AND ( ( ( Grb2 ) ) OR ( ( Shc ) ) ) ) OR ( NIK AND ( ( ( Shc ) ) OR ( ( Grb2 ) ) ) ) OR ( Grb2 AND ( ( ( Shc ) ) ) ) OR ( Ras ) OR ( Trafs ) OR ( Rac ) OR ( GCK ) OR ( Cdc42 )
Palpha_qR = ( alpha_qR AND ( ( ( GRK ) ) ) )
DOCK180 = ( Crk AND ( ( ( Cas ) ) AND ( ( PIP3_345 ) ) ) )
MLK1 = ( Cdc42 ) OR ( Rac )
Ras = ( RasGRF_GRP ) OR ( SHP2 ) OR ( Sos )
Gbg_i = ( alpha_iR AND ( ( ( NOT Gbg_i ) ) AND ( ( NOT Gai ) ) ) ) OR ( Gai )
Nck = ( Cas ) OR ( EGFR )
RKIP = ( PKC )
MKK7 = ( Mekk4 AND ( ( ( ASK1 ) ) ) ) OR ( MLK1 AND ( ( ( ASK1 ) ) ) ) OR ( MLK2 AND ( ( ( ASK1 ) ) ) ) OR ( MLK3 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk1 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk3 AND ( ( ( ASK1 ) ) ) )
Erk = ( Mek ) OR ( ( ( Erk ) AND NOT ( MKPs ) ) AND NOT ( PP2A ) )
MLK2 = ( Cdc42 AND ( ( ( SAPK ) ) ) ) OR ( Rac AND ( ( ( SAPK ) ) ) )
MKK3 = ( Mekk4 AND ( ( ( ASK1 ) ) ) ) OR ( MLK1 AND ( ( ( ASK1 ) ) ) ) OR ( MLK2 AND ( ( ( ASK1 ) ) ) ) OR ( MLK3 AND ( ( ( ASK1 ) ) ) ) OR ( TAK1 AND ( ( ( ASK1 ) ) ) ) OR ( Tpl2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk3 AND ( ( ( ASK1 ) ) ) ) OR ( PAK AND ( ( ( ASK1 ) ) ) ) OR ( TAO_12 AND ( ( ( ASK1 ) ) ) )
Src = ( ( Gas AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( PTPa ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( alpha_sR AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( Fak AND ( ( ( PTP1b ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( Cas AND ( ( ( PTP1b ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( Gai AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( EGFR ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) )
DAG = ( ( PLC_B AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( DGK AND ( ( ( DAG ) ) ) ) ) OR ( ( PLC_g AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( DGK AND ( ( ( DAG ) ) ) ) ) OR ( DAG AND ( ( ( NOT DGK ) ) ) )
RalGDS = ( ( ( alpha_sR AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Ras AND ( ( ( PDK1 ) ) AND ( ( PIP3_345 ) ) ) ) ) AND NOT ( PKC ) ) OR ( ( ( alpha_qR AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Ras AND ( ( ( PDK1 ) ) AND ( ( PIP3_345 ) ) ) ) ) AND NOT ( PKC ) ) OR ( ( ( alpha_iR AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Ras AND ( ( ( PDK1 ) ) AND ( ( PIP3_345 ) ) ) ) ) AND NOT ( PKC ) ) OR ( ( ( alpha_1213R AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Ras AND ( ( ( PDK1 ) ) AND ( ( PIP3_345 ) ) ) ) ) AND NOT ( PKC ) )
RGS = ( CaM AND ( ( ( PIP3_345 ) ) ) )
RalBP1 = ( Ral )
CaMKK = ( CaM )
EGFR = ( EGF AND ( ( ( NOT PKC ) ) ) ) OR ( alpha_iR AND ( ( ( Ca AND PKC ) ) ) ) OR ( alpha_qR AND ( ( ( Ca AND PKC ) ) ) ) OR ( alpha_1213R AND ( ( ( Ca AND PKC ) ) ) )
RhoGDI = ( NOT ( ( AA ) OR ( PKC ) OR ( PIP2_45 ) ) ) OR NOT ( PKC OR PIP2_45 OR AA )
|
Global control of cell cycle transcription by coupled CDK and
network oscillators
David A. Orlando1,2, Charles Y. Lin1, Allister Bernard3, Jean Y. Wang1, Joshua E. S.
Socolar4, Edwin S. Iversen5, Alexander J. Hartemink3, and Steven B. Haase1,*
1Department of Biology, Duke University
2Program in Computational Biology and Bioinformatics, Duke University
3Department of Computer Science, Duke University
4Department of Physics, Duke University
5Department of Statistical Science, Duke University
Abstract
A significant fraction of the Saccharomyces cerevisiae genome is transcribed periodically during the
cell division cycle1,2, suggesting that properly timed gene expression is important for regulating cell
cycle events. Genomic analyses of transcription factor localization and expression dynamics suggest
that a network of sequentially expressed transcription factors could control the temporal program of
transcription during the cell cycle3. However, directed studies interrogating small numbers of genes
indicate that their periodic transcription is governed by the activity of cyclin-dependent kinases
(CDKs)4. To determine the extent to which the global cell cycle transcription program is controlled
by cyclin/CDK complexes, we examined genome-wide transcription dynamics in budding yeast
mutant cells that do not express S-phase and mitotic cyclins. Here we show that a significant fraction
of periodic genes were aberrantly expressed in the cyclin mutant. Surprisingly, although cells lacking
cyclins are blocked at the G1/S border, nearly 70% of periodic genes continued to be expressed
periodically and on schedule. Our findings reveal that while CDKs play a role in the regulation of
cell cycle transcription, they are not solely responsible for establishing the global periodic
transcription program. We propose that periodic transcription is an emergent property of a
transcription factor network that can function as a cell cycle oscillator independent of, and in tandem
with, the CDK oscillator.
The biochemical oscillator controlling periodic events during the cell cycle is centered on the
activity of cyclin-dependent kinases (CDKs) (reviewed in ref. 5). The cyclin/CDK oscillator
governs the major events of the cell cycle, and in embryonic systems this oscillator functions
in the absence of transcription, relying only on maternal stockpiles of mRNAs and proteins.
CDKs are also thought to act as the central oscillator in somatic cells and yeast, and directed
*Corresponding author Mailing address: DCMB Group, Dept. of Biology Box 90338 Science Drive Durham, NC 27708-0338 Phone:
919.613.8205 Fax: 919.613.8177 shaase@duke.edu.
Author Contributions D.A.O., C.Y.L., and S.B.H. designed and performed the experiments. J.Y.W. provided technical expertise.
D.A.O., C.Y.L., A.B., E.S.I., and A.J.H. performed the computational analyses, with contributions from J.E.S.S and S.B.H. to the Boolean
model. D.A.O. and S.B.H. prepared the manuscript with contributions from C.Y.L., A.B., E.S.I., and A.J.H.
Supplementary Information is linked to the online version of the paper at www.nature.com/nature.
Author Information The microarray data discussed in this publication have been deposited in NCBI’s Gene Expression Omnibus (GEO,
http://www.ncbi.nlm.nih.gov/geo/) and are accessible through GEO series accession number GSE8799. Reprints and permissions
information is available at www.nature.com/reprints. Correspondence and requests for materials should be addressed to S.B.H
(shaase@duke.edu).
NIH Public Access
Author Manuscript
Nature. Author manuscript; available in PMC 2009 September 2.
Published in final edited form as:
Nature. 2008 June 12; 453(7197): 944–947. doi:10.1038/nature06955.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
studies suggest that they play an essential role in controlling the temporally ordered program
of transcription (reviewed in refs. 4,6). However, systems-level analyses using high throughput
technologies1,2,7,8 have suggested alternative models for regulation of periodic transcription
during the yeast cell cycle1,3,9. By correlating genome-wide transcription data with global
transcription factor binding data, models have been constructed in which periodic transcription
is an emergent property of a transcription network1,3,9. In these networks, transcription factors
expressed in one cell cycle phase bind to the promoters of genes encoding transcription factors
that function in a subsequent phase. Thus, the temporal program of transcription could be
controlled by sequential waves of transcription factor expression, even in the absence of
extrinsic control by cyclin/CDK complexes.
The validity and relevance of the intrinsically oscillatory transcription factor network
hypotheses remain uncertain, because for the limited number of periodic genes that have been
dissected in detail, periodic transcription was found to be governed by CDKs (reviewed in ref.
4). Thus, we sought to determine to what extent CDKs and transcription factor networks
contribute to global regulation of the cell cycle transcription program. To this end, we
investigated the dynamics of genome-wide transcription in budding yeast cells disrupted for
all S-phase and mitotic cyclins (clb1,2,3,4,5,6). These cyclin mutant cells are unable to replicate
DNA, separate SPBs, undergo isotropic bud growth, or complete nuclear division, indicating
that they are devoid of functional Clb/CDK complexes10-12. So by conventional cell cycle
measures, clb1,2,3,4,5,6 cells arrest at the G1/S border. We have previously shown that
clb1,2,3,4,5,6 cells cyclically trigger G1 events10, including the activation of G1-specific
transcription and bud emergence. Nevertheless, if Clb/CDK activities are essential for
triggering the transcriptional program, then periodic expression of S-phase and G2/M-specific
genes should not be observed.
We examined global transcription dynamics in synchronized populations of both wild-type
cells and cyclin mutant cells. Synchronous populations of early G1 cells were collected by
centrifugal elutriation. Cell aliquots were then harvested at 16 min intervals for 270 min
(equivalent to ~2 cell cycles in wild-type and ~1.5 in the cyclin mutant). Transcript levels were
measured genome-wide for each time point using Yeast 2.0 oligonucleotide arrays (Affymetrix,
Santa Clara, CA). Results from two independent experiments each for both wild-type and
cyclin mutant cells were highly reproducible, with adjusted r2 values of 0.995 and 0.989,
respectively (Supplementary Fig. 1). All statistical analyses were performed using replicate
data sets, but to facilitate illustration, single data sets were used for all graphical representations.
To identify periodically transcribed genes, we applied a modification of the method developed
by de Lichtenberg et al.13 to data acquired from our wild-type cells. We established a set of
1271 genes that were transcribed periodically (Fig. 1a, and Supplementary Table 1). This set
of periodic genes shares 510 and 577 genes with those sets previously identified as periodic
by Spellman et al.2 and Pramila et al.1, respectively (Supplementary Fig. 2), with 440
consensus periodic genes identified by all three studies (Supplementary Table 2). We then
examined the transcriptional dynamics of our set of 1271 periodic genes in the cyclin mutant
(Fig. 1b). The behavior of many genes changed significantly in the cyclin mutant, supporting
previous findings. However, despite the fact that cyclin mutant cells arrest at the G1/S border,
a large fraction of periodic genes in all cell cycle phases continued to be expressed on schedule
(Fig. 1b). Similar cyclin-dependent and -independent behaviors are also observed in the set of
440 consensus periodic genes (Supplementary Fig. 3).
Using absolute change and Pearson correlation analyses (see Supplementary Information), we
determined that 833 of the periodic genes exhibited changes in expression behavior in the cyclin
mutant and thus are likely to be directly or indirectly regulated by B-cyclin/CDK.
Orlando et al.
Page 2
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Our genome-level experiments accurately reproduced previous findings regarding several
well-studied B-cyclin/CDK-regulated genes (Fig. 2). We observed that a subset of late G1
transcripts (SBF-regulated genes like CLN2 but not MBF-regulated genes like RNR1) were not
fully repressed (Fig. 2a and b) as expected in mitotic cyclin mutant cells 14,15. A subset of M/
G1 transcripts (including SIC1 and NIS1), are targets of the transcription factors Swi5 and
Ace2, which are normally excluded from the nucleus by CDK phosphorylation until late
mitosis16-19. SIC1 and NIS1 were expressed earlier in the cyclin mutant (Fig. 2c and d)
presumably because nuclear exclusion of Swi5 and Ace2 is lost in cyclin mutant cells. The
modest degree of shift in the timing of SIC1 and NIS1 transcription likely reflects the fact that
SWI5 and ACE2 transcripts do not accumulate to maximal levels in cyclin mutant cells as
expected for Clb2 cluster genes (including CDC20) (Fig. 2e and f) 14,20,21. Although a
significant fraction of periodic genes exhibited changes in the amplitude of expression
(increased or decreased), a statistical analysis of the dynamic range of expression across all
periodic genes revealed that the majority of genes in cyclin mutant cells exhibit only modest
changes, if any, with respect to wild-type cells (Supplementary Fig. 4).
To identify novel subsets of co-regulated genes based on transcriptional behaviors observed
in both wild-type and cyclin mutant cells, we employed the affinity propagation algorithm22
to first cluster genes based on expression in wild-type cells, and then subcluster genes based
on their behavior in cyclin mutant cells (Fig. 3 and Supplementary Fig. 5). Of the 833 cyclin-
regulated genes, 513 were assigned to 30 discrete clusters exhibiting similar behaviors in wild-
type cells (Fig. 3a, and Supplementary Fig. 6), and were then subclustered into 56 novel clusters
based on their transcription profiles in cyclin mutant cells (Fig. 3b and Supplementary Table
3). Using data from global transcription factor localization studies23, we identified subsets of
transcription factors that may regulate these subclusters using an over-representation analyses
(Fig. 3 and Supplementary Table 4). Based on their association with the promoters of genes in
cyclin-regulated subclusters, these factors are likely to be directly or indirectly regulated by
cyclins. Consistent with this hypothesis, several of these factors have already been shown to
be CDK targets14,15,18,19,24-29. These findings lay the groundwork for elucidating the full
range of mechanisms by which cyclin/CDKs regulate transcription during the cell cycle.
Strikingly, 882 of the genes identified as periodic in wild-type cells, continued to be expressed
on schedule in cyclin mutant cells despite cell cycle “arrest” at the G1/S border (Fig. 4a and
b). Some of these genes (450 in total) exhibited minor changes in transcript behavior but
continued to be expressed at the proper time, as shown above for ACE2. Thus, some genes that
were cyclin-regulated are also included in the set of genes that maintain periodicity.
Nevertheless, a statistical analysis of the dynamic range of expression of these genes in wild-
type and cyclin mutant cells indicates that the amplitude changes for most of these genes is
quite modest (see Supplementary Figs. 7, 8 and 9). The finding that nearly 70% of the genes
identified as periodic in wild-type cells are still expressed on schedule in cyclin mutant cells
demonstrates the existence of a cyclin/CDK-independent mechanism that regulates temporal
transcription dynamics during the cell cycle. This observation is supported by the analysis of
the set of 440 consensus periodic genes, the bulk of which maintain periodicity in the cyclin
mutant (Supplementary Fig. 10).
In principle, a transcription network defined by sequential waves of transcription factor
expression1,3,9 might function independent of any extrinsic control by CDKs. To determine
if a transcription network could account for cyclin/CDK-independent periodic transcription,
we constructed a synchronously updating Boolean network model and determined that such a
model can indeed explain the periodic expression patterns we observed in cyclin mutant cells
(Fig. 4c). Transcription factors that maintained periodicity in the cyclin mutant were placed
on a circularized cell cycle time line based on their peak time of transcription in the cyclin
mutant. Connections were drawn based on documented physical interactions23,30
Orlando et al.
Page 3
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
(Supplementary Table 5) between a transcription factor and the promoter region of a gene
encoding a transcription factor expressed subsequently (see Supplementary Information). The
architecture of the network in cyclin mutant cells is virtually identical to that in wild-type cells
(Supplementary Fig. 11), and is also remarkably similar to models based on wild-type
expression data from previous studies1,3,9.
When the network is endowed with Boolean logic functions (Supplementary Table 6a),
synchronous updating of the model leads to a cycle that produces successive waves of
transcription by progressing through five distinct states before returning to the initial state
(Supplementary Fig. 12a and b). Thus, the model functions as an oscillator and produces a
correctly-sequenced temporal program of transcription.
To examine the robustness of the network oscillator, we evaluated outcomes when initializing
the network from all possible starting states. Over 80% of the 512 starting states entered the
oscillatory cycle depicted in Fig. 4c, with the remainder terminating in a steady state where all
genes were transcriptionally inactive (Supplementary Table 6b and c). We also examined
whether the oscillations were sensitive to the choice of the Boolean logic functions assigned
to nodes with multiple inputs, specifically, the activating inputs to Cln3 and SFF, and the
repressors of SBF and Cln3. For most of the logic functions, the predominant outcome was
again the oscillatory cycle depicted in Fig. 4c, but in some cases, the model enters two
qualitatively similar cycles (Supplementary Fig. 12c and d, and Supplementary Table 6), with
the remainder again terminating in a transcriptionally inactive steady state. Several Boolean
logic functions produce the same cycles (Supplementary Table 6b), so the model cannot
precisely determine the true logic of the network connections. Nevertheless, the fact that the
model can produce qualitatively similar cycles, and that these cycles can be reached from many
initial states, suggests that robust oscillation is an emergent property of the network
architecture.
Previous studies proposed that a cyclin/CDK-independent oscillator could trigger some
periodic events, including bud emergence10. The robust oscillating character of our model
suggests that a transcription factor network may function as this cyclin/CDK-independent
oscillator. Because cyclin genes are themselves among the periodic genes targeted by this
network, and because cyclin/CDKs can, in turn, influence the behavior of transcription factors
in the network, precise cell cycle control could be achieved by coupling a transcription factor
network oscillator with the cyclin/CDK oscillator. The existence of coupled oscillators could
explain why the cell cycle is so robust to significant perturbations in gene expression or cyclin/
CDK activity.
Our findings also suggest that the properly scheduled expression of genes required for cell
cycle regulated processes such as DNA synthesis and mitosis is not sufficient for triggering
these events. The execution of cell cycle events in wild-type cells is likely to require both
properly timed transcription and post-transcriptional modifications mediated by CDKs.
Methods Summary
Strains and cell synchronization
Wild-type and cyclin mutant strains of S. cerevisiae are derivatives of BF264-15Dau, and were
constructed by standard yeast methods. The clb1,2,3,4,5,6 GAL1-CLB1 mutant strain along
with growth conditions and synchrony procedures was described previously10,11.
RNA isolation and microarray analysis
Total RNA was isolated time points (every 16 min for a total of 15 time points) as described
previously10. mRNA was amplified and fluorescently labeled using GeneChip One-Cycle
Orlando et al.
Page 4
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Target Labeling (Affymetrix, Santa Clara, CA) . Hybridization to Yeast 2.0 oligonucleotide
arrays (Affymetrix, Santa Clara, CA) and image collection were performed at the Duke
Microarray Core Facility (http://microarray.genome.duke.edu/) according to standard
Affymetrix protocols.
Data analysis
A workflow diagram for data analysis is depicted in Supplementary Fig. 13.
Methods
Strains and cell synchronization
Yeast strains were grown in rich YEP medium (1% yeast extract, 2% peptone, 0.012% adenine,
0.006% uracil) containing 2% galactose. Forty-five minutes prior to elutriation, dextrose was
added to YEP 2% galactose medium to terminate CLB1 expression from the GAL1 promoter.
After elutriation, wild-type and clb1,2,3,4,5,6 GAL1-CLB1 cells were grown in rich YEP 2%
dextrose,1M sorbitol at 30°C at a density of 107/ml. Sorbitol was added to stabilize cells with
elongated buds. Aliquots of 50ml (cell density = 107/ml) were harvested every 8 min for 4hr.
Budding index was determined microscopically by counting ≥ 200 cells for each time point.
Data analysis
CEL files from all 60 oligonucleotide arrays were normalized, and summarized using the dChip
method32 as implemented in the affy package (v.1.8.1) within Bioconductor using default
parameters. The output of this package is a measure of absolute expression levels for each
probe in arbitrary expression units (Fig. 2). Data presented in heat maps and centroid line graphs
(Figs. 1, 3, and 4, and Supplementary Figs. 3, 5, 8, 11,12, 15-18) are expressed as log2-fold
change for each gene relative to its mean expression over the interval from the first G1 to the
second S phase.
The CLOCCS population synchrony model31 was used to temporally align expression data
from our two wild-type and two cyclin mutant experiments. Briefly, the CLOCCS model allows
the alignment of data from multiple synchrony/time series experiments to a common cell cycle
time line using budding as a parameter measured on single cells at each time point31. Although
cyclin mutant cells arrest at the G1/S border by conventional measures, G1 events, such as bud
emergence, are activated periodically with a cycle time similar to wild-type cells10. Thus, the
CLOCCS model can be used to temporally align cycles in wild-type and cyclin mutant cells.
Because the kinetics of synchrony/release experiments can vary, and wild-type and cyclin
mutant cells have marginally different cycle times, alignment is imperative for meaningful
comparison of data. We used the CLOCCS parameter estimates to align all four data sets such
that the population level measurements were mapped onto a common cell cycle timeline. The
timeline utilizes standard cell cycle phases (as determined by measured parameters) and an
additional phase (Gr) corresponding to a period of recovery from the initial synchrony
procedure that overlaps with early G131. The recovery period (Gr) was eliminated from most
of the data displayed, as the genes expressed in this period tend to be specific to this period
and are not expressed again in the next cell cycle in either wild-type or cyclin mutant cells.
The CLOCCS model was designed for wild-type yeast populations but can accommodate data
from the cyclin mutant with minor modifications (see Supplementary Information). CLOCCS
model fits for both the wild-type and cyclin mutant datasets are shown in Supplementary Fig.
14, and the corresponding parameter estimates are shown in Supplementary Table 7.
A modification of the method described by de Lichtenberg13 was used to determine the subset
of genes exhibiting periodic transcription (see Supplementary Information for details). The
methods used to identify genes with altered transcriptional profiles (Fig. 3), and similar profiles
Orlando et al.
Page 5
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
(Figs. 4a and b) in cyclin mutant cells with respect to wild-type cells are also described in detail
in Supplementary Information. Methods for determining over-represented transcription factors
in the clusters (Fig. 3) and details regarding the construction of the synchronously updating
Boolean network model (Fig. 4c) can also be found in Supplementary Information. Two
additional analyses similar to that performed in Fig. 3 were performed on consensus periodic
genes (Supplemental Table 2) identified as changing expression as well as those identified as
maintaining periodicity. Details and results of those analyses can be found in Supplementary
Information (Supplementary Figs. 15-19 and Supplementary Tables 8 and 9)
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgments
We would like to thank D. Lew and L. Simmons Kovacs for helpful discussions and critical reading of the manuscript,
and P. Benfey for helpful discussions and support. We gratefully acknowledge financial support from the American
Cancer Society (to S.B.H.), Alfred P. Sloan Foundation (to A.J.H.), National Science Foundation (to A.J.H. and
J.E.S.S.), and National Institutes of Health (to S.B.H., A.J.H., and J.E.S.S.).
References
1. Pramila T, Wu W, Miles S, Noble WS, Breeden LL. The Forkhead transcription factor Hcm1 regulates
chromosome segregation genes and fills the S-phase gap in the transcriptional circuitry of the cell
cycle. Genes Dev 2006;20:2266–78. [PubMed: 16912276]
2. Spellman PT, et al. Comprehensive identification of cell cycle-regulated genes of the yeast
Saccharomyces cerevisiae by microarray hybridization. Mol Biol Cell 1998;9:3273–97. [PubMed:
9843569]
3. Simon I, et al. Serial regulation of transcriptional regulators in the yeast cell cycle. Cell 2001;106:697–
708. [PubMed: 11572776]
4. Wittenberg C, Reed SI. Cell cycle-dependent transcription in yeast: Promoters, transcription factors,
and transcriptomes. Oncogene 2005;24:2746–55. [PubMed: 15838511]
5. Murray AW. Recycling the cell cycle: Cyclins revisited. Cell 2004;116:221–34. [PubMed: 14744433]
6. Zwicker J, Muller R. Cell cycle-regulated transcription in mammalian cells. Prog Cell Cycle Res
1995;1:91–9. [PubMed: 9552355]
7. Cho RJ, et al. A genome-wide transcriptional analysis of the mitotic cell cycle. Mol Cell 1998;2:65–
73. [PubMed: 9702192]
8. Ren B, et al. Genome-wide location and function of DNA binding proteins. Science 2000;290:2306–
9. [PubMed: 11125145]
9. Lee TI, et al. Transcriptional regulatory networks in Saccharomyces cerevisiae. Science 2002;298:799–
804. [PubMed: 12399584]
10. Haase SB, Reed SI. Evidence that a free-running oscillator drives G1 events in the budding yeast cell
cycle. Nature 1999;401:394–7. [PubMed: 10517640]
11. Haase SB, Winey M, Reed SI. Multi-step control of spindle pole body duplication by cyclin-dependent
kinase. Nature Cell Biology 2001;3(1):38–42.
12. Lew DJ, Reed SI. Morphogenesis in the yeast cell cycle: Regulation by Cdc28 and cyclins. J. Cell
Biol 1993;120:1305–1320. [PubMed: 8449978]
13. de Lichtenberg U, et al. Comparison of computational methods for the identification of cell cycle-
regulated genes. Bioinformatics 2005;21:1164–71. [PubMed: 15513999]
14. Amon A, Tyers M, Futcher B, Nasmyth K. Mechanisms that help the yeast cell cycle clock tick: G2
cyclins transcriptionally activate G2 cyclins and repress G1 cyclins. Cell 1993;74:993–1007.
[PubMed: 8402888]
15. Koch C, Schleiffer A, Ammerer G, Nasmyth K. Switching transcription on and off during the yeast
cell cycle: Cln/Cdc28 kinases activate bound transcription factor SBF (Swi4/Swi6) at start, whereas
Orlando et al.
Page 6
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Clb/Cdc28 kinases displace it from the promoter in G2. Genes Dev 1996;10:129–41. [PubMed:
8566747]
16. Toyn JH, Johnson AL, Donovan JD, Toone WM, Johnston LH. The Swi5 transcription factor of
Saccharomyces cerevisiae has a role in exit from mitosis through induction of the CDK-inhibitor
Sic1 in telophase. Genetics 1997;145:85–96. [PubMed: 9017392]
17. Knapp D, Bhoite L, Stillman DJ, Nasmyth K. The transcription factor Swi5 regulates expression of
the cyclin kinase inhibitor p40SIC1. Mol Cell Biol 1996;16:5701–7. [PubMed: 8816483]
18. Moll T, Tebb G, Surana U, Robitsch H, Nasmyth K. The role of phosphorylation and the CDC28
protein kinase in cell cycle-regulated nuclear import of the S. cerevisiae transcription factor, SWI5.
Cell 1991;66:743–758. [PubMed: 1652372]
19. O’Conallain C, Doolin MT, Taggart C, Thornton F, Butler G. Regulated nuclear localisation of the
yeast transcription factor Ace2p controls expression of chitinase (CTS1) in Saccharomyces
cerevisiae. Mol Gen Genet 1999;262:275–82. [PubMed: 10517323]
20. Zhu G, et al. Two yeast forkhead genes regulate the cell cycle and pseudohyphal growth. Nature
2000;406:90–4. [PubMed: 10894548]
21. Kumar R, et al. Forkhead transcription factors, Fkh1p and Fkh2p, collaborate with Mcm1p to control
transcription required for M-phase. Curr Biol 2000;10:896–906. [PubMed: 10959837]
22. Frey BJ, Dueck D. Clustering by passing messages between data points. Science 2007;315:972–6.
[PubMed: 17218491]
23. Harbison CT, et al. Transcriptional regulatory code of a eukaryotic genome. Nature 2004;431:99–
104. [PubMed: 15343339]
24. Costanzo M, et al. CDK activity antagonizes Whi5, an inhibitor of G1/S transcription in yeast. Cell
2004;117:899–913. [PubMed: 15210111]
25. de Bruin RA, et al. Constraining G1-specific transcription to late G1 phase: the MBF-associated
corepressor Nrm1 acts via negative feedback. Mol Cell 2006;23:483–96. [PubMed: 16916637]
26. Ho Y, Costanzo M, Moore L, Kobayashi R, Andrews BJ. Regulation of transcription at the
Saccharomyces cerevisiae start transition by Stb1, a Swi6-binding protein. Mol Cell Biol
1999;19:5267–78. [PubMed: 10409718]
27. Pic-Taylor A, Darieva Z, Morgan BA, Sharrocks AD. Regulation of cell cycle-specific gene
expression through cyclin-dependent kinase-mediated phosphorylation of the forkhead transcription
factor Fkh2p. Mol Cell Biol 2004;24:10036–46. [PubMed: 15509804]
28. Sidorova JM, Mikesell GE, Breeden LL. Cell cycle-regulated phosphorylation of Swi6 controls its
nuclear localization. Mol Biol Cell 1995;6:1641–58. [PubMed: 8590795]
29. Ubersax JA, et al. Targets of the cyclin-dependent kinase Cdk1. Nature 2003;425:859–64. [PubMed:
14574415]
30. Teixeira MC, et al. The YEASTRACT database: A tool for the analysis of transcription regulatory
associations in Saccharomyces cerevisiae. Nucleic Acids Res 2006;34:D446–51. [PubMed:
16381908]
31. Orlando DA, et al. A probabilistic model for cell cycle distributions in synchrony experiments. Cell
Cycle 2007;6:478–88. [PubMed: 17329975]
32. Li C, Wong WH. Model-based analysis of oligonucleotide arrays: model validation, design issues
and standard error application. Genome Biol 2001;2:0032.1–0032.11.res
Orlando et al.
Page 7
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Figure 1.
Dynamics of periodic transcripts in wild-type and cyclin mutant cells. Heat maps depicting
mRNA levels of periodic genes are shown for a, wild-type and b, cyclin mutant cells. Each
row in a and b represents data for the same gene (Supplementary Table 1). Transcript levels
are expressed as log2-fold change vs. mean expression. Transcript levels at each point in the
time series were mapped onto a cell cycle timeline31 (see Methods). The S and G2/M phases
of the cyclin mutant timeline are shaded, indicating that by conventional definitions, cyclin
mutant cells arrest at the G1/S-phase transition.
Orlando et al.
Page 8
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Figure 2.
Transcription dynamics of established cyclin/CDK-regulated genes. Absolute transcript levels
(dChip-normalized Affymetrix intensity units/1000) are shown for the SBF- and MBF-
regulated genes a, CLN2 and b, RNR1; the Ace2/Swi5-regulated genes c, SIC1 and d, NIS1;
and the Clb2 cluster genes e, CDC20 and f, ACE2. Wild-type cells (solid line) and cyclin mutant
cells (dashed line).
Orlando et al.
Page 9
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Figure 3.
Genes exhibiting altered behaviors in cyclin mutant cells. a, Clusters of genes with similar
expression patterns in wild-type cells. b, Subclusters of genes with similarly altered expression
patterns in cyclin mutant cells. Each row in a and b represents data for the same gene
(Supplementary Table 1). Transcript levels are depicted as shown in Fig. 1. Up to five over-
represented transcription factors for each cluster are shown (see Methods and Supplementary
Table 4 for complete lists).
Orlando et al.
Page 10
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Figure 4.
The periodic transcription program is largely intact in cyclin mutant cells that arrest at the G1/
S border. a, Genes maintaining periodic expression in cyclin mutant cells exhibit similar
dynamics in b, wild-type cells. Each row in a and b represents the same gene (Supplementary
Table 1). Transcript levels are depicted as shown in Fig. 1. c, Synchronously updating Boolean
network model. Transcription factors are arranged based on the time of peak transcript levels
in cyclin mutant cells. Arrows indicate transcription factor/promoter interaction. Activating
interactions, outer rings; repressive interactions, inner rings. Coloring indicates activity in one
of five successive states; SBF and YHP1 are active in two states (Supplementary Table 6).
Orlando et al.
Page 11
Nature. Author manuscript; available in PMC 2009 September 2.
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
|
18463633
|
MBF = ( CLN3 )
HCM1 = ( MBF AND ( ( ( SBF ) ) ) )
SWI5 = ( SFF )
YOX1 = ( MBF AND ( ( ( SBF ) ) ) )
SFF = ( SBF AND ( ( ( HCM1 ) ) ) )
ACE2 = ( SFF )
YHP1 = ( MBF ) OR ( SBF )
SBF = ( ( ( MBF ) AND NOT ( YHP1 ) ) AND NOT ( YOX1 ) ) OR ( ( ( CLN3 ) AND NOT ( YHP1 ) ) AND NOT ( YOX1 ) )
CLN3 = ( ( ( SWI5 AND ( ( ( ACE2 ) ) ) ) AND NOT ( YOX1 ) ) AND NOT ( YHP1 ) )
|
BioMed Central
Page 1 of 15
(page number not for citation purposes)
BMC Systems Biology
Open Access
Research article
A logic-based diagram of signalling pathways central to macrophage
activation
Sobia Raza1, Kevin A Robertson1,3, Paul A Lacaze1, David Page1,
Anton J Enright2, Peter Ghazal*1,3 and Tom C Freeman*1
Address: 1Division of Pathway Medicine, University of Edinburgh, The Chancellor's Building, College of Medicine, 49 Little France Crescent,
Edinburgh, EH16 4SB, UK, 2Computation and Functional Genomics Laboratory, Sanger Institute, Wellcome Trust Genome Campus, Hinxton,
Cambridge, CB10 1SA, UK and 3Centre for Systems Biology, University of Edinburgh, Darwin Building, King's Building Campus, Mayfield Road,
Edinburgh, EH9 3JU, UK
Email: Sobia Raza - sobia.raza@ed.ac.uk; Kevin A Robertson - kevin.robertson@ed.ac.uk; Paul A Lacaze - paul.lacaze@ed.ac.uk;
David Page - david.page@ed.ac.uk; Anton J Enright - aje@sanger.ac.uk; Peter Ghazal* - p.ghazal@ed.ac.uk;
Tom C Freeman* - tfreeman@staffmail.ed.ac.uk
* Corresponding authors
Abstract
Background: The complex yet flexible cellular response to pathogens is orchestrated by the
interaction of multiple signalling and metabolic pathways. The molecular regulation of this response
has been studied in great detail but comprehensive and unambiguous diagrams describing these
events are generally unavailable. Four key signalling cascades triggered early-on in the innate
immune response are the toll-like receptor, interferon, NF-?B and apoptotic pathways, which co-
operate to defend cells against a given pathogen. However, these pathways are commonly viewed
as separate entities rather than an integrated network of molecular interactions.
Results: Here we describe the construction of a logically represented pathway diagram which
attempts to integrate these four pathways central to innate immunity using a modified version of
the Edinburgh Pathway Notation. The pathway map is available in a number of electronic formats
and editing is supported by yEd graph editor software.
Conclusion: The map presents a powerful visual aid for interpreting the available pathway
interaction knowledge and underscores the valuable contribution well constructed pathway
diagrams make to communicating large amounts of molecular interaction data. Furthermore, we
discuss issues with the limitations and scalability of pathways presented in this fashion, explore
options for automated layout of large pathway networks and demonstrate how such maps can aid
the interpretation of functional studies.
Background
The innate immune response is executed at the molecular
level by a complex series of interwoven signalling path-
ways. In this context, pathways may be defined as a net-
work of directional interactions between the components
of a cell which orchestrate an appropriate shift in cellular
activity in response to a specific biological input or event.
Whilst our ability to perform quantitative and qualitative
measurements on the cellular components has increased
massively in recent years, as has our knowledge on how
Published: 23 April 2008
BMC Systems Biology 2008, 2:36
doi:10.1186/1752-0509-2-36
Received: 23 January 2008
Accepted: 23 April 2008
This article is available from: http://www.biomedcentral.com/1752-0509/2/36
© 2008 Raza et al; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 2 of 15
(page number not for citation purposes)
they interact with each other, we still struggle to translate
these observations into graphical and computationally
tractable models. However without such models we can
not hope to truly understand biology at a systems level.
Traditionally, representations of molecular pathways have
been produced ad hoc and frequently included in reviews
and original papers. Whilst they are clearly useful aids to
understanding cellular events, even at their best, they are
not sufficient by themselves, relying on extensive textual
descriptions to explain what is shown pictorially. Recent
years have seen considerable growth in the availability of
public and commercial databases offering searchable
access to pathways and interaction data derived from a
combination of manual and automated (text mining)
extraction of primary literature, reviews and large-scale
molecular interaction studies. Using these tools it is pos-
sible to view a range of canonical pathway views or gener-
ate networks of interactions based on a given query.
However, all of these efforts are let down by one or a
number of key factors. The notation used in diagrams to
depict one molecule's interaction with another is varied,
often ambiguous and therefore limited in its ability to
depict the exact nature of the relationship between com-
ponents of a pathway. There is often a lack of direct access
to the experimental evidence relating to the interactions
depicted or to the dataset as a whole. Similarly, labelling
of the pathway components often uses non-standard
nomenclature or mixes protein names from one species
with that of another, such that again the reader is left
uncertain as to what exactly is being shown. Finally, path-
way diagrams usually focus only on a small part of a bio-
logical system and one which often reflects the curator's
bias, such that the 'same' pathway described by different
individuals may share little in common. Whatever the
source of these pathways and networks they generally suf-
fer from graphically poor representation with ambiguity
around the precise identity of what is being shown and
the exact nature of their interaction. In order to address
these issues the groups of Kohn and Kitano began to
devise new approaches to pathway notation using many
ideas adopted from the electronics industry [1-3]. In par-
ticular the MIM (molecular interaction map) notation [3]
a form of entity-relationship representation and the proc-
ess description notation (PDN) [1], respectively. Since
then there has been an increasing interest in the systems
biology community to develop a consensus view on a
standard approach for representing biological pathways
[4]. Whilst this process is now well advanced there is cur-
rently no internationally agreed standard graphical nota-
tion system for building pathway diagrams and a paucity
of worked examples of this type of notation in use. Exam-
ples of pathways that have been published using these
notation systems include a molecular interaction map of
macrophage signalling [5] and Toll-Like-Receptor signal-
ling [6] which have been depicted using the PDN scheme
and cell cycle control and DNA repair presented in the
MIM notation [2].
Over the last four years we have been developing a nota-
tion scheme for the depiction of biological pathways that
borrows many of the ideas of existing notation systems
but attempts to address some of their short comings. The
Edinburgh Pathway Notation [26] uses a logical state-
transition representation to describe biological pathways,
similar to PDN. The work described here follows on from
this initial publication and reports a modified version of
the EPN scheme which is aligned with the developing
international SBGN standard but has a number of impor-
tant differences with the scheme as currently proposed.
Crucially, the notation provides a logical context for inter-
actions between components in the pathway, it can dis-
play the temporal order of reactions and can be mapped
to the machine-readable SBML (systems biology markup
language) [7]. Of primary importance to this notation
scheme and indeed the SBGN is the desire to develop
pathway maps that are 'readable' by a biologist. Since the
pathway maps are primarily produced as a tool for com-
munication it is critical that they are easily understanda-
ble and the notation can be applied and read by biologists
with minimal training. Other objectives (of the SBGN) are
that the notation should be computable, compact, show
sub-cellular localization and be tolerable of incomplete
knowledge. Whilst all of these objectives are valid, fulfill-
ing them in practice is far from trivial and there are few
worked examples of large pathway diagrams depicted in
standard notations, available in the public domain.
The innate immune response is orchestrated by series of
signalling pathways that have evolved to elicit an appro-
priate defensive response to attack by pathogenic organ-
isms. Pathogen sensing involves pattern recognition
receptors such as the toll-like receptors (TLR's) which in
mammalian cells constitutes a family of up to 11 [8] trans-
membrane receptors each responsible for distinguishing
particular
pathogen-associated
molecular
patterns
(PAMPs). Detection of pathogen molecules by these
receptors results in the recruitment of various adaptor pro-
teins and the activation of downstream signal transduc-
tion cascades [9,10]. Activation of de novo gene expression
follows, which ultimately acts to recruit new proteins and
augment the response to infection. Interferons (IFNs) are
central to this response, as are (amongst others) interferon
regulatory factors (IRFs), JAK/STAT signalling proteins
and the nuclear factor-kappa B (NF-?B) family of proteins
[11]. The IRF family of transcription factors bind specific
DNA sequences, as do the STAT proteins, present on the
promoter of target genes [12,13]. NF-?B signalling can reg-
ulate transcription through a combination of NF-?B pro-
tein homo- and heterodimers [14-17]. These pathways are
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 3 of 15
(page number not for citation purposes)
also known to regulate components of the apoptotic path-
way, thereby providing the potential for cells to undergo
a programmed cell death [18], the ultimate cellular sacri-
fice in defence of the organism.
The TLR, IFN, NF-?B and apoptosis pathways are of central
importance in defining the macrophages response to
pathogens and do so in a highly inter-dependant manner
[11]. Extensive literature describing the pathways and
their interconnectivity, like so many others in biology, is
available but only from multiple and disparate sources. In
our effort to understand these events as a basis for inter-
preting analyses of host-pathogen interactions and the
inflammatory response in the macrophage, we have
endeavoured to construct an integrated and logic-based
pathway diagram of signalling cascades fundamental to
macrophage activation using a current version of the EPN
scheme. We present the results of these labours as an
example of our on going work in this area and hope that
this map will be used to supplement and contrast with the
efforts of others [6] in this area.
Methods
Collection of Molecular Interaction Data and Biological
Representation of Pathways
In an effort to describe and consolidate knowledge of
pathways central to macrophage activation we have con-
structed a pathway diagram based on published literature.
Ideally, two published papers citing protein-protein or
protein-gene interactions were required for the inclusion
of a given interaction on to the pathway diagram. In some
circumstances we accepted one piece of published evi-
dence if the paper described extensive experimental verifi-
cation of the interaction. This was deemed necessary as
two publications per interaction can limit inclusion of
potentially interesting interactions included in other
pathway resources (KEGG, Reactome etc) and newly dis-
covered interactions. It is also important to note that the
primary task of this exercise was to develop a 'consensus'
of knowledge and information about a given pathway.
A list of interactions to be mapped was compiled [see
Additional file 1], including details about the nature of
the interaction and source of the information. A pathway
map was then drawn using the principles laid down by the
EPN scheme. These include the concept that the molecu-
lar components of a pathway be they proteins, protein
complexes and genes (or in principle any other cellular
component that plays a part in a pathway) are represented
as simple shapes containing a unique and unambiguous
identifying label. Attempts to depict pictorially the func-
tional activity or functional domains of components have
been avoided as this adds to the visual complexity of the
diagram and can be misleading. For consistency compo-
nents (nodes) have been named by their official human
genome nomenclature (HGNC) symbol, although in cer-
tain instances we have felt it necessary due to the wide-
spread use of other naming conventions to supplement
this with additional annotation. For example we have
used the name tBID to differentiate the truncated (active)
form of the protein from its precursor (BID) and similarly
in order to distinguish the native (inactive) form of cas-
pases we used the suffix Pro e.g. ProCASP3 from the active
cleaved form (CASP3). We have also included additional
naming conventions to differentiate between protein
forms e.g. in the NF-?B pathway (p50, p52 etc) or
included common aliases where they are prevalent in the
literature, these names being placed in brackets after the
official name. Whilst the use of such ad hoc naming con-
ventions is in theory undesirable, they are still in common
use and alternative ways to differentiate between protein
forms is not supported under the HGNC and standard
naming conventions for describing proteins in their vari-
ous modified forms (truncated, cleaved, activated by
cleavage etc) does not yet exist. Where pathway compo-
nents are protein complexes, the name of the complex is
given as a concatenation of the names of its constituent
parts, although this has in some cases been supplemented
by the inclusion of common names such as 'apoptosome'
to denote the complex between CASP9, CYCS and APAF1.
Components are depicted at the site of their activity and
are shown only once in any given cellular compartment
unless different activation states of the components are
known due to phosphorylation, ubquitinisation, cleavage
etc., when these molecular states may be shown as con-
nected but individual entities. The state of a component
may be shown as a supplement to the components name
e.g. active [A], inactive [I], phosphorylated [P]. Interac-
tions (edges) between components or transitions between
one cellular compartment and another, are shown as
arrows which either contact interacting partners via
Boolean logic operators (&, OR, NOT) and/or transition/
annotation nodes that provide information as to the
nature of the interaction or transition from one state to
another. Attempts to depict molecular details of interac-
tions and state transitions such as the exact site of a pro-
tein's phosphorylation, have generally been avoided.
Whilst important, if depicted on a map of this size the
information quickly clutters up the diagram rendering it
inaccessible to the casual reader. However, in cases where
such details are necessary to differentiate one component
form from another they should be added. Finally, layout
of the elements and interactions that make up the path-
way should be such that it is relatively easy to follow the
direction and nature of flow of information from the ini-
tial trigger to the eventual outcome. In an effort to achieve
this, where possible interacting map components are
drawn close together keeping edge lengths short and easy
to follow, crossover of edges is kept to a minimum and
every effort is taken to keep connecting edges separate,
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 4 of 15
(page number not for citation purposes)
with a minimum number of changes in direction to get
from one point to another.
The pathway map was drawn using the freely available
program yEd graph editor (yFiles software, Tubingen, Ger-
many). yEd is a general purpose graphical tool designed
for the depiction of networks. Although not specifically
designed for biological pathway depiction it has been
used previously for this and similar purposes [19,20] and
has a range of characteristics and capabilities that make it
ideally suited for the job. Initially pathways were laid out
by hand. Areas of the canvas were defined as representing
specific compartments of the cell e.g. plasma membrane,
cytoplasm, nucleus etc., and cellular components and the
interactions in which they took part were drawn in the
appropriate space. A section of the overall map describing
IFNG receptor signalling laid out according to the cellular
location of the components has been included as an
example of the notation scheme in action (Figure 1) and
a list of notation symbols used here is provided in Figure
2.
The combined map of macrophage activation pathways
described here (Figure 3) is available for download [21]
and presented in a number of image (.jpeg, .pdf) and
graphical formats (.xml, .graphml). The .graphml file [see
Additional file 2] can be opened in yEd graph editor [22]
and in this format is available for editing or expansion.
PubMed IDs supporting the interactions of the pathway
are stored on appropriate edges within the .graphml ver-
sion of the diagram. We have found the yEd program to
be relatively intuitive to use and to require minimal or no
training. Hence the pathway diagram presented here is
easily accessible, distributable and can be modified by
end users to suit their interests or knowledge-base. The
EPN can be mapped to SBML and we are in the process of
creating a SBML version of the map described here.
As the complexity of maps increases and the interactions
between components become evermore intertwined,
manual organization of these events becomes time-con-
suming and difficult. However, the pathway diagrams
have been specifically drawn as directional networks. As
such layout of the pathway maps can be aided by use of
various automated layout algorithms. The hierarchical
layout and classic-orthogonal edge routing applications
within the yEd software were the most effective in terms of
providing an easily interpretable view of directional flow
in the diagram (Figure 4a, b). However, other layout algo-
rithms such as the organic layout function (Figure 4c) can
also provide different views of the pathway. Furthermore
this pathway can be easily converted into a 3-D network
(Figure 4d) in BioLayout Express3D [23]. Whilst 3-D path-
way networks do not readily support user readability of
Manual layout of Type II interferon signalling (IFNG) taken from the integrated pathway diagram (Figure 2)
Figure 1
Manual layout of Type II interferon signalling (IFNG) taken from the integrated pathway diagram (Figure 2).
The pathway is arranged to flow from left to right. Components are coloured according to type (protein, complex or gene)
and arranged within the sub-cellular compartments in which they are active. This pathway is initiated by IFNG binding to its
receptor and a subsequent phosphorylation cascade involving a number of the JAK and STAT family of proteins. Several tran-
scriptionally active complexes are formed (STAT1 homodimer, ISGF3 complex, STAT1:STAT1:IRF9 complex) and the pathway
culminates with the transcriptional activation of target genes.
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 5 of 15
(page number not for citation purposes)
the interactions, it provides an environment where very
large graphs may be plotted (15,000 nodes, 2.5 million
edges) and queried. As such these tools can aid interpreta-
tion of the innate structure within the network of interac-
tions of large pathway diagrams and together provide a
solution, albeit not necessarily a perfect one, to the issue
of scalability. With these capabilities it will be possible to
scale up these diagrams to the point where they may con-
tain thousands of components, operators and transition
nodes.
Network Analysis of the Transcriptional Response of
Mouse Bone Marrow Derived Macrophages to Interferon-
gamma Treatment
Primary mouse bone marrow derived monocytes were
prepared from male balb/c mice 10–12 weeks old. Cells
were washed, resuspended in DMEM-F12/10% FCS/L929
medium and counted before being plated in a 24-well
plate at a concentration of 5 × 105 cells/well. To differen-
tiate the cells from monocytes into primary macrophages,
cells were then incubated for 7 days in DMEM-F12 growth
media supplemented with 10% L929 cell suspension
releasing the MCP-1 macrophage stimulating factor, with
media changes on days 3 and 5. On day 7 the growth
medium was replaced with DMEM-12/10%FCS medium
containing 10 u/ml recombinant mouse interferon-
gamma (Pierce-Thermofisher Scientific, Rockford US) and
harvested 1, 2, 4 & 8 h following treatment or collected
pre-treatment (0 h). Total RNA was harvested from the
cells using an RNeasy Plus kit (Qiagen) according to man-
ufacturer's instructions. RNA was quantified and quality
controlled using a NanoDrop spectrophotometer (Nano-
Symbols of the modified Edinburgh Pathway Notation
Figure 2
Symbols of the modified Edinburgh Pathway Notation. Unique shapes and identifiers are used to distinguish between
each element of the notation allowing its interpretation even in the absence of colour. Colour maybe used for aesthetic pur-
poses and to ease identification of nodes. The notation can be broadly divided into four categories; components, boolean oper-
ators, transition nodes and annotated edges. Components consist of any interacting species from proteins, complexes, genes
or other molecular species (pathogens, DNA, RNA). Pathway initiators are also presented in the notation. Boolean operators
are essential for capturing the dependencies of an interaction. Transition nodes provide information as to the nature of the
interaction (such as cleavage, translocation, phosphorylation). Edges are directional and can be coloured for visual impact. Dis-
tinctive arrow-heads are used to distinguish between the pathway inputs and outputs but are otherwise avoided. Instead in-line
edge annotation is used to add a visual cue as to the meaning of an edge. Cellular compartmental information is provided by
physical location and backdrop or by colouring nodes according to their sub-cellular location.
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 6 of 15
(page number not for citation purposes)
Integrated pathway map of signalling in the macrophage
Figure 3
Integrated pathway map of signalling in the macrophage. The diagram includes the interferon signalling, NF-?B, apop-
tosis and toll-like receptor pathways, all represented as one integrated pathway due to their overlapping interactions. In gen-
eral interactions of the interferon response pathway are in the top quarter of the map, with NF-?B directly below. Apoptosis is
presented halfway down the map and toll-like receptor signalling is in the bottom quarter. 154 different protein or gene nodes
are included in the pathway, along with 80 different complexes and 12 other molecular species (such as pathogens, DNA,
RNA). The pathway diagram represents 272 different interactions.
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 7 of 15
(page number not for citation purposes)
Automated layouts of the pathway diagram
Figure 4
Automated layouts of the pathway diagram. (4a) a hierarchical-classic layout was applied to the entire pathway and the
orientation was set to flow from left to right. Nodes are coloured according to their sub-cellular location. With this layout the
flow of pathway information and biological logic is maintained, such that the inputs to pathway are placed at the left side of the
diagram and these can be followed through to the outputs at the right hand side. (4b) a detailed inset of the hierarchical-classic
layout of the integrated pathway taken from 4a. (4c) Organic-classic automated layout of the entire pathway generated in yEd
graph editor. Although the directionality of flow in the pathway is lost, interacting partners tend to be placed in close proximity
of each other in this layout. (4d) a 3-dimentional network of the apoptosis interactions in the pathway generated using BioLay-
out Express3D. This network can be queried for pathway information. Unique shapes are used to identify the different pathway
notation symbols; spheres denote interacting components (proteins, genes, complexes), decahedron shapes represent boolean
operators or transition nodes and tetrahedron shapes correspond to the in-line edge annotation (in this case activation, or
inhibition). All notation symbols are coloured to correspond to the colour scheme applied in the 2-dimentional pathway dia-
gram (e.g. complexes are yellow, proteins are blue, and activation-annotations are green). Furthermore interacting compo-
nents are sized according to type, such that spheres representing complexes appear larger than proteins or genes. As with the
2-dimentional diagram the colour scheme used is customisable. The 3-dimentional network retains the information captured in
the 2-dimentional pathway and although spatial placement of nodes in relation to their sub-cellular location has been lost, this
information can be retrieved by querying the network and/or colouring nodes according to their sub-cellular location.
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 8 of 15
(page number not for citation purposes)
Drop Technologies) and BioAnalyser 2100 (Agilent). Rep-
licate 150 ng samples of total RNA derived from two
separate wells per time point were labelled using the
Affymetrix whole transcript labelling protocol and hybrid-
ized for 16 h at 45°C to Affymetrix mouse exon 1.0 ST
arrays. They were then washed and scanned according to
manufacturer's recommendations.
Data (ArrayExpress Ac. No: E-MEXP-1490) was normal-
ized using the RMA package within the Affymetrix Expres-
sion Console software and annotated. Transcripts which
might be considered to be differentially expressed were
identified using either the Empirical Bayes function
within Bioconductor [24] or using the annova function
within GeneSpring (Agilent Technologies, Stockport,
Cheshire) with a 1.6 fold cut-off. In total 1,678 transcripts
were identified by one or both of these filters. The data
corresponding to this list was then loaded into the net-
work visualization tool BioLayout Express3D [23] using a
Pearson correlation cut-off of 0.9 to filter edges. The
resultant network graph (Figure 5a) of 1,491 nodes was
clustered using the graph-based clustering algorithm MCL
[25] set at an inflation value of 2.2 resulting in 26 clusters
(Figure 5a & b). Clusters composed of transcripts that
were up-regulated were then further collated into 3
groups; genes up-regulated at (1) 1–2 hours, (2) 2–4
hours and (3) 4–8 hours post-treatment and genes that
were both differently expressed and present on the inte-
grated pathway diagram were highlighted on the map.
Results and Discussion
We set out to use the EPN scheme as originally published
[26]. However, during construction of the maps described
here the notation system was found to be too limiting to
convey certain biological concepts and overly compli-
cated for others. A simplification of certain aspects of the
notation was therefore deemed necessary in order to
achieve the objectives outlined above, in particular
human readability. Modifications made to the EPN were
not intended to change the built in logic of the notation
scheme but rather merely enhance the visual characteris-
tics of the diagrams produced. One of the major modifica-
a) A network graph of differentially expressed genes following Ifng treatment
Figure 5
a) A network graph of differentially expressed genes following Ifng treatment. A Pearson correlation cut-off of 0.9
was set to filter edges in the network and the resultant graph was clustered using the graph-based clustering algorithm MCL
set at an inflation value of 2.2. Each node represents a transcript and nodes are coloured according to the cluster to which they
belong. Nodes belonging to the same cluster share a common pattern of expression over the time-course following Ifng treat-
ment. b) A view of the 26 clusters defined from the network graph in 5a. The size of each sphere representing a clus-
ter corresponds to the size of its node membership. Clusters are assigned a description of the co-expression pattern they
present over the time course and are coloured according to whether the nodes within those clusters are up-regulated
(orange) or down-regulated (green) following the Ifng treatment.
a
b
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 9 of 15
(page number not for citation purposes)
tions we have made is in the reliance of the original EPN
(and the emerging SBGN standard) on multiple types of
arrow heads to infer different meaning to the interactions.
We have used only one type of arrowhead and relied far
more heavily on the use of transition nodes or annotation
nodes to infer the nature of the transition from one
molecular state to another and add information to edges.
We found this system to improve the readability of the
maps as well as provide greater flexibility in the range of
concepts that may be depicted. The pathway diagrams cre-
ated using this notation scheme function without the use
of colour and do not therefore lose their semantics if
viewed without it. Nevertheless, colour does provide a
powerful device for increasing the visual impact of the fig-
ure. Here we have generally chosen apposite or symbolic
colours to represent the appropriate interaction; for exam-
ple red for inhibition, green for activation. However, it
must be emphasized that the exact colour scheme is not
important and should be seen as customizable to suit an
individuals taste or limitations in colour recognition.
The pathway map described here (Figure 3) consists of a
total of 295 nodes of which 140 are proteins, 99 com-
plexes, 44 genes, and 12 other components (pathogens,
DNA, RNA etc). A total of 272 interactions are described
in the pathway map, of these 85 are binding events, 149
are various activation state modulations (67 activation of
gene expression, 26 phosphorylation, 7 auto-phosphor-
ylation, 1 dephosphoylation, 23 cleavage, 9 transloca-
tions and 16 activation by processes not defined). There
are 10 inhibition reactions, 4 of these are inhibition of
gene expression, 3 are inhibition of cleavage, and 1 is an
inhibition of translocation. A total of 26 translocation
events occur as well as 2 protein dissociations. 282 differ-
ent references support the interactions shown on the path-
way [see Additional file 3]. In many circumstances the
same paper may describe multiple interactions, for exam-
ple Chaudhary et al., (1997) report that both TNFRSF10A
and TNFRSF10B recruit the protein FADD during apopto-
sis signalling [27]. A detailed description of the biological
content of this pathway diagram is given in Additional file
4.
In order to check the integrity of the network each input
(e.g. cytokine or pathogen molecule), was highlighted in
turn and the logical flow of information from this input
followed through the diagram. By following the flow of
information from each pathway input, a different but
expected output was observed, be that the activation of
transcription or a process such as apoptosis (Figure 6a &
b). This suggests that although several signalling pathways
have been integrated to form this diagram the specificity
of connectivity has not been lost.
Use of Pathway Diagram in the Interpretation of
Transcriptomics Data
In order to demonstrate the utility of this pathway dia-
gram in the interpretation of transcriptomics data we have
examined the transcriptional events following the treat-
ment of mouse bone marrow derived macrophages
(BMDM) with interferon-gamma (Ifng). Using the net-
work analysis tool BioLayout Express3D [23] we con-
structed a 3-D network of transcripts identified as being
differently expressed following Ifng stimulation (Figure
5a). 1,491 transcripts were represented within the net-
work, 1,274 of which grouped into 26 clusters with ? 5
members (Figure 5b). There are 154 unique proteins/
genes represented on the pathway map, 55 of which are
represented within these clusters and a further 3 compo-
nents were in the transcriptional network but did not fall
into a cluster [see Additional file 5]. All of the genes repre-
sented on the map were in clusters of up-regulated genes.
Clusters of transcripts representing genes activated at dif-
ferent times following treatment were then further col-
lated into 3 groups of up-regulated genes; genes activated
at (1) 1–2 hours, (2) 2–4 hours and (3) 4–8 hours post-
treatment. Genes that were activated and included in the
set of mapped genes were then highlighted on the map
and the possible downstream consequences (assuming de
novo protein synthesis and activity following an increase
in gene transcription) were highlighted (Figure 7). In this
way is has been possible for the first time to interpret these
transcriptional events in the context of the possible conse-
quences of these observations.
During the very early phase (0–2 hours) of the response to
Ifng treatment only two genes, SOC3 and IER3, corre-
sponded to pathway components shown in the diagram
(Figure 7a). SOCS3 (suppressor of cytokine signalling 3)
is an inhibitory protein of Interferon-gamma receptor
complex signalling and has also been reported elsewhere
to be expressed in macrophages following interferon treat-
ment [28]. The up-regulation of SOCS3 represents a clas-
sical negative feedback loop required to regulate the
magnitude and duration of signalling downstream of the
IFNG receptor signalling, in addition to limiting the
response to any subsequent cytokine stimulus [29,30].
IER3 (immediate early response 3) a stress inducible gene
is a target gene of the NF-?B signalling complex NFKB1-
RELA [31] and is known to be activated in response to a
variety of cellular stress signals [32-35]. Although IER3 is
not depicted to be directly induced by Jak-Stat signalling
we understand that connectivity exists between this sig-
nalling system and the NF-?B pathway. 25 components of
the pathway diagram were also regulated 2–4 hours post-
Ifng treatment (Figure 7b). Most noticeably members of
apoptosis and TLR signalling were changing during this
time and interestingly these changes occurred around the
initiation or receptor signalling region of these pathways.
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 10 of 15
(page number not for citation purposes)
Follow through of signalling pathways stimulated by IFNG (6a) and FASLG (6b)
Figure 6
Follow through of signalling pathways stimulated by IFNG (6a) and FASLG (6b). The signalling events following the
input signals of IFNG and FASLG have been highlighted on the entire map in lilac and orange, respectively. The nodes activated
or directly affected by FASLG or IFN-gamma binding to their receptors are coloured and the interaction edges and gates are
also highlighted. Nodes and edges not directly downstream of the FASLG or IFNG signalling are shown in grey. This figure
demonstrates inputs into the pathway can clearly be followed to the expected outcome events. In the case of IFNG-input, gene
transcription is the resulting event, and in the case of FASLG, apoptosis. Furthermore these examples clearly depict the inter-
actions of the pathway can be followed logically and do not result in unexpected crosstalk.
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 11 of 15
(page number not for citation purposes)
When observed in more detail we identified that three
potential mechanisms of apoptosis induction were tar-
geted; TNF, TNFRSF10 and FAS signalling. TNF, its recep-
tor TNFRSF1A and an adaptor protein RIPK1 are all up-
regulated, as is TNFSF10 (Trail-ligand). FAS and adaptor
molecules (DAXX and CFLAR) of the FAS receptor were
also increased in their expression. A similar observation
was also made for TLR-signalling, as a number of key
adaptors proteins (including MYD88 and IRAK2) were
up-regulated in the 2–4 hour timeframe. By activating the
TLR system and apoptotic machinery the cells appear to
preparing themselves for contact with pathogens and
priming themselves for apoptosis. One possible conse-
quence of TLR signalling when followed though on the
pathway diagram is the activation of the IRF5 transcrip-
tion factor and indeed 5 targets of IRF5 were up-regulated
at the 2–4 hour time phase (CXCL11, IFIT1, CXCL10,
IFIT2, and TNFSF10). Moreover IRF5 was itself regulated
at the later time points (4–8 hours) post-IFNG treatment.
Another consequence of TLR-signalling is the activation of
the NF-?B pathway and again the key constituents of this
pathway (NFKB1 and RELA) were activated at the later
time points as were some transcriptional targets of this
complex. During the 4–8 hours period BID, an important
amplifier of apoptotic input signals via the mitochondrial
apoptotic pathway, was up-regulated (Figure 7c). BID can
be cleaved and activated by any of the three aforemen-
tioned apoptotic mechanisms (FASLG, TNFSF10 and
TNF) [36-38] that were altered during the earlier time
phase. Also up during the latter hours were members of
the Jak-Stat pathway (JAK2, STAT1, STAT2, and PRKCD
which phosphorylates and activates STAT1) and some tar-
get genes of the Jak-Stat pathway, which could represent
increased sensitivity to IFN or other cytokine signalling.
The integrated pathway diagram presented at (a) 1–2 hours, (b) 2–4 hours and (c) 4–8 hours post-Ifng treatment
Figure 7
The integrated pathway diagram presented at (a) 1–2 hours, (b) 2–4 hours and (c) 4–8 hours post-Ifng treat-
ment. Differentially expressed genes are highlighted in red and the possible consequential downstream events resulting from
the changes, (assuming de novo protein synthesis) are highlighted in blue.
SOCS3
IER3
NFKBIA
IL15
CXCL10
IFIT2
TNF
TNFRSF1A
BCL3
CXCL11
IFIT1
TNFSF10
RIPK1
DAXX
CFLAR
CASP4
LMNA
RIPK3
MYD88
IRAK2
TLR3
TNFSF10
STAT2
STAT1
JAK2
IRF2
GBP1
TAP1
CIITA
IRF2
CCL5
ICAM1
CD40
RELA
NFKB1
TBK1
BIRC3
SOD2
TRAF2
TRAF1
NFKB2
IRF5
BID
TLR2
TLR6
0-2 hours
2-4 hours
4-8 hours
a
TLR9
b
c
SOCS1
IRF1
IRF8
HIST2H4
FAS
STAT1
PSMB9
CXCL9
PRKCD
TICAM1
TRAF2
CASP1
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 12 of 15
(page number not for citation purposes)
We are acutely aware that the current pathway diagram
covers only a relatively small number of the genes shown
to be transcriptionally regulated following Ifng treatment.
For instance none of the genes shown to be down-regu-
lated by Ifng are shown in the diagram. However even
with the current limited coverage we have been able to
extrapolate some interesting observations by visualizing
the changes and the possible downstream effect of the
changes. It has been possible to appreciate the connectiv-
ity and co-dependency of the changes over time and using
this approach the detail of how signalling in one region
may have downstream effects on another signalling sys-
tem can be hypothesized and in many examples here
extracted.
Critical Review of Pathway
In constructing this integrated map of macrophage activa-
tion pathways we have attempted to represent events in a
detailed, accurate and logical fashion. However, it must be
emphasized that this map is by its nature a biased view of
events. Its construction has been primarily driven by our
interest in understanding signalling events in the macro-
phage and interpretation of the literature is an unavoida-
bly flawed process; determining what constitutes good
evidence for an interaction and what does not, is often dif-
ficult to judge especially for those who do not specifically
work in the area. Furthermore, any view of what consti-
tutes a given pathway is also highly subjective and is
always being driven by an individual's perspective and sci-
entific trends, as well as current knowledge. Even though
pathway diagrams typically depict individual pathways in
isolation of other systems, in reality it is well recognized
that there is significant overlap in pathway membership
and cross-talk between related pathways. Input from one
signalling pathway can influence the outcome in another,
underscoring the need to view the connections between
various signalling systems. Indeed, when one searches for
the known interactions of any well characterized protein
using database tools such as String [39] or Ingenuity [40]
one is potentially led in many directions, each interacting
protein in turn leading to an ever expanding network of
molecular interactions. Therefore when drawing pathway
maps such as the one described here, it is impossible to
include all the known interactions of any given compo-
nent. We are aware there are other systems important in
their regulation which have could be included, most
noticeably, NOD/NALP receptor signalling, MAP kinase
cascades, interleukin and other cytokine/chemokine sys-
tems, many aspects of the TNF-family of proteins, antigen
presentation and cell cycle pathway, to name but a few.
Some of these systems are now being added to the path-
way diagram but this is largely being driven by our need
to interpret the results of systems-level analysis of the
macrophages response to pathogens and cytokines.
Indeed, the fact that this pathway is far from complete is
further emphasized by its use in interpreting the transcrip-
tional response to Ifng. Of the 1,141 transcripts falling
into clusters of co-regulated genes following Ifng treat-
ment, only 55 were represented on the map and the map
so far includes only 44 genes in total as being regulated by
any transcription factor. This therefore highlights the fact
that there is some considerable way to go if we are to gen-
erate a complete model of the potential downstream
events following the interferon signalling cascade.
In the case of the signalling systems described here, the
interaction data is derived from the available literature
and is therefore dependant on the quality of that work,
the biological system from which that information was
derived and as already mentioned represents a subjective
view of the information available. Seldom do signalling
pathways operate independently of each other therefore
analyzing only a subset of nodes known to belong to a
particular pathway is unlikely to be insightful as to the
activity of the system as a whole. With so many of pieces
of the jigsaw missing and many aspects of the activity of
these large integrated molecular networks still unknown,
performing meaningful analyses on relatively small sec-
tions of what is otherwise an immense network of inter-
acting proteins, is unlikely to deliver accurate or
biologically representative predictions for some time.
The current notation system used for the pathway pre-
sented here arguably works well up to this size of pathway
and the end result we hope will serve as a useful reference
for biologists interested in these systems. However, scala-
bility of pathway diagrams is an important issue especially
when a compromise must be reached between presenting
a human readable map with one that captures the exten-
sive interaction data now available for many molecules.
Although we intend to continue to consolidate and add
interactions to the current map we are aware that this
could prove difficult in number of respects. When new
components are added, in order to place them near to the
site of their interacting partners the layout of the entire
graph sometimes needs to be manually altered to make
space. Furthermore, as functional units of an integrated
pathway network frequently share components, proteins
often referred to as hubs, it is often impossible to place a
component near to all its interacting partners requiring
edges (interactions) to span large distances across the
map. One method of reducing long edge lengths is to
depict individual components more than once within a
given cellular compartment. However, this in turn adds to
the issue of scalability as the additional nodes consume
more space, add more complexity and the visual link
between components of the pathway are lost. We have
therefore been exploring alternative approaches to over-
come the issue of scalability in pathway depictions. One
approach is to use automated layout algorithms to draw
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 13 of 15
(page number not for citation purposes)
the relationships between pathway components. Certain
layout algorithms are very effective at displaying connec-
tivity between components with little or no need for man-
ual intervention (Figure 4a & b). This allows the rendering
of relatively large pathway diagrams quickly and easily,
whilst retaining much of the biologist friendly aspects to
the diagrams. What is lost is the spatial layout according
to the cellular compartment of components. However this
aspect can be retained, at least in part, by the use of colour
to signify in which compartment they reside. A second
approach for dealing with large interaction networks/
pathways is to visualize them in 3-dimensional space.
Using the tool BioLayout Express3D recently developed by
us [23] we have found it possible to render very large net-
works. In this instance the shape, size and colour can all
be used to distinguish between different component types
and colour can be overlaid to indicate cellular compart-
ment (Figure 4d). Whilst arrow heads are not supported in
3-D mode directionality is reinstated when graphs or
selected portions of large graphs are converted to 2-D net-
works.
Conclusion
With the majority of the components of life defined, at
least at some level, there is an increasing desire to put the
parts together in order to construct models of biological
systems which can be tested and refined. In this respect,
the value of logically presented pathway diagrams is
becoming ever more apparent given the growing need to
systematically organize and describe the interactions
between the various components that make up a cell.
Pathway diagrams serve several purposes; they can be
used to capture a large amount of information, provide a
point of reference for researchers with an interest in the
pathway or particular member of that pathway, and can
be used to aid the interpretation of systems level analyses.
The pathway presented here is by no means a comprehen-
sive view of all the pathways involved in macrophage acti-
vation, but acts a worked example of how a number of key
pathways might be represented in what we hope is a logi-
cal and unambiguous fashion. However, with the visual
modifications to the EPN scheme we believe we have ful-
filled the primary objectives of providing a graphical nota-
tion that is both useable by biologists and which could
still serve as the basis for computational model develop-
ment. So whilst others have gone some way to address the
issue of human readability of their pathway diagrams we
believe that we have derived an elegant yet simple nota-
tion scheme that better addresses the needs of biologists.
The mapping process is a continuing effort and during the
next steps we aim to consolidate and expand the content
of the diagram. This in turn may require refinements to
the notation system as issues in depicting the relation
between components and the cellular components in
which they are active arise. As we enhance our under-
standing of individual signalling pathways and how they
integrate with others this will aid understanding of immu-
nological disorders at a molecular level. Building pathway
diagrams or networks of interactions from the existing
knowledgebase is one of the milestones towards the appli-
cation of pathway and systems biology to the field of
medicine.
Authors' contributions
SR constructed the integrated pathway diagram, contrib-
uted to the development of the notation system, partici-
pated in the expression profiling and analysis, and helped
to draft the manuscript. KR contributed to the pathway
development efforts and standardisation of pathway data
collection and storage. PL contributed to the construction
of pathway, collection of molecular interaction data and
development of the notation scheme. DP set up the Ifng
time course study. AE developed BioLayout Express3D vis-
ualisation of the pathway diagram. PG originally con-
ceived the EPN scheme and has supported its continued
development. TF oversaw and contributed to the pathway
construction, orchestrated the development of the EPN
scheme, conceived the Ifng time course study, and its
analysis and drafted the manuscript.
Additional material
Additional file 1
Pathway interaction list. Interactions included in the pathway map are
listed (in no particular order) in this data file. Official HGNC (human
gene nomenclature committee) gene symbols are used to name the inter-
acting components along with a brief description of the type of interaction
and its cellular location. Entrez gene IDs of interacting components are
also provided as are the PubMed IDs of the reference(s) supporting each
interaction.
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-2-36-S1.xls]
Additional file 2
Diagram of signalling pathways central to macrophage activation. This
file can be opened, viewed and edited by users using the freely available
graph-editor software yEd (yFiles software, Tubingen, Germany). This
can be downloaded at [22] where full downloading instructions are
described. PubMed IDs supporting the interactions of the pathway are
stored on appropriate edges within this .graphml yEd file. Once an edge is
selected the PubMed ID may be viewed within the descriptions tab of the
properties box for that edge.
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-2-36-S2.zip]
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 14 of 15
(page number not for citation purposes)
Acknowledgements
This work was supported in part by INFOBIOMED EU FP6 programme,
BBSRC and Wellcome Trust.
References
1.
Kitano H, Funahashi A, Matsuoka Y, Oda K: Using process dia-
grams for the graphical representation of biological net-
works. Nat Biotechnol 2005, 23(8):961-966.
2.
Kohn KW: Molecular interaction map of the mammalian cell
cycle control and DNA repair systems. Mol Biol Cell 1999,
10(8):2703-2734.
3.
Kohn KW, Aladjem MI, Weinstein JN, Pommier Y: Molecular inter-
action maps of bioregulatory networks: a general rubric for
systems biology. Mol Biol Cell 2006, 17(1):1-13.
4.
Systems Biology Graphical Notation [http://sbgn.org/]
5.
Oda K, Kimura T, Matsuoka Y, Funahashi A, Muramatsu M, Kitano H:
Molecular interaction map of a macrophage. AfCS Research
Reports 2004, 2(14):12.
6.
Oda K, Kitano H: A comprehensive map of the toll-like recep-
tor signaling network. Mol Syst Biol 2006, 2:2006 0015.
7.
Hucka M, Finney A, Sauro HM, Bolouri H, Doyle JC, Kitano H, Arkin
AP, Bornstein BJ, Bray D, Cornish-Bowden A, Cuellar AA, Dronov S,
Gilles ED, Ginkel M, Gor V, Goryanin, Hedley WJ, Hodgman TC,
Hofmeyr JH, Hunter PJ, Juty NS, Kasberger JL, Kremling A, Kummer
U, Le Novere N, Loew LM, Lucio D, Mendes P, Minch E, Mjolsness
ED, Nakayama Y, Nelson MR, Nielsen PF, Sakurada T, Schaff JC, Sha-
piro BE, Shimizu TS, Spence HD, Stelling J, Takahashi K, Tomita M,
Wagner J, Wang J: The systems biology markup language
(SBML): a medium for representation and exchange of bio-
chemical network models. Bioinformatics 2003, 19(4):524-531.
8.
Takeda K, Akira S: Toll-like receptors in innate immunity. Int
Immunol 2005, 17(1):1-14.
9.
Kagan JC, Medzhitov R: Phosphoinositide-mediated adaptor
recruitment controls Toll-like receptor signaling. Cell 2006,
125(5):943-955.
10.
Fitzgerald KA, Chen ZJ: Sorting out Toll signals. Cell 2006,
125(5):834-836.
11.
Zhong B, Tien P, Shu HB: Innate immune responses: crosstalk
of signaling and regulation of gene transcription. Virology 2006,
352(1):14-21.
12.
Levy DE, Kessler DS, Pine R, Reich N, Darnell JE Jr.: Interferon-
induced nuclear factors that bind a shared promoter ele-
ment correlate with positive and negative transcriptional
control. Genes Dev 1988, 2(4):383-393.
13.
Darnell JE Jr., Kerr IM, Stark GR: Jak-STAT pathways and tran-
scriptional activation in response to IFNs and other extracel-
lular signaling proteins. Science 1994, 264(5164):1415-1421.
14.
Chen FE, Huang DB, Chen YQ, Ghosh G: Crystal structure of p50/
p65 heterodimer of transcription factor NF-kappaB bound
to DNA. Nature 1998, 391(6665):410-413.
15.
Cramer P, Larson CJ, Verdine GL, Muller CW: Structure of the
human NF-kappaB p52 homodimer-DNA complex at 2.1 A
resolution. Embo J 1997, 16(23):7078-7090.
16.
Ghosh G, van Duyne G, Ghosh S, Sigler PB: Structure of NF-kappa
B p50 homodimer bound to a kappa B site. Nature 1995,
373(6512):303-310.
17.
Huang DB, Huxford T, Chen YQ, Ghosh G: The role of DNA in
the mechanism of NFkappaB dimer formation: crystal struc-
tures of the dimerization domains of the p50 and p65 subu-
nits. Structure 1997, 5(11):1427-1436.
18.
Kerr JF, Wyllie AH, Currie AR: Apoptosis: a basic biological phe-
nomenon with wide-ranging implications in tissue kinetics.
Br J Cancer 1972, 26(4):239-257.
19.
Butte AJ, Kohane IS: Creation and implications of a phenome-
genome network. Nat Biotechnol 2006, 24(1):55-62.
20.
Singh A, Mai D, Kumar A, Steyn AJ: Dissecting virulence pathways
of Mycobacterium tuberculosis through protein-protein
association. Proc Natl Acad Sci U S A 2006, 103(30):11346-11351.
21.
Division of Pathway Medicine, University of Edinburgh.
www.pathwaymedicine.ed.ac.uk .
22.
yWorks
-
The
Diagramming
Company.
http://
www.yworks.com/en/index.html .
23.
Freeman T.C. GL Brosch M., van Dongen S., Mazière P., Grocock R.J,
Freilich S., Thornton J. and Enright A.J.: Construction, Visualisa-
tion and Clustering of Transcription Networks from Micro-
array Expression Data. PLoS Computational Biology 2007 in press.
24.
Bioconductor open source software for bioinformatics
[http://www.bioconductor.org/]
25.
van Dongen S: Graph clustering by flow simulation. [PhD disser-
tation] Utrecht (The Netherlands): University of Utrecht 2000:169.
26.
Moodie SL SA Goryanin I, Ghazal P: A Graphical Notation to
Describe the Logical Interactions of Biological Pathways.
Journal of Integrative Bioinformatics 2006, 3(2):11.
27.
Chaudhary PM, Eby M, Jasmin A, Bookwalter A, Murray J, Hood L:
Death receptor 5, a new member of the TNFR family, and
DR4 induce FADD-dependent apoptosis and activate the
NF-kappaB pathway. Immunity 1997, 7(6):821-830.
28.
Gil MP, Bohn E, O'Guin AK, Ramana CV, Levine B, Stark GR, Virgin
HW, Schreiber RD: Biologic consequences of Stat1-independ-
ent IFN signaling. Proc Natl Acad Sci U S A 2001, 98(12):6680-6685.
29.
Alexander WS, Hilton DJ: The role of suppressors of cytokine
signaling (SOCS) proteins in regulation of the immune
response. Annu Rev Immunol 2004, 22:503-529.
30.
Kubo M, Hanada T, Yoshimura A: Suppressors of cytokine sign-
aling and immunity. Nat Immunol 2003, 4(12):1169-1176.
31.
Huang YH, Wu JY, Zhang Y, Wu MX: Synergistic and opposing
regulation of the stress-responsive gene IEX-1 by p53, c-Myc,
and multiple NF-kappaB/rel complexes. Oncogene 2002,
21(44):6819-6828.
32.
Charles CH, Yoon JK, Simske JS, Lau LF: Genomic structure,
cDNA sequence, and expression of gly96, a growth factor-
Additional file 3
Bibliography of references supporting interactions on the integrated path-
way diagram. Within this document a list of the 282 different references
supporting the interactions on the pathway map are provided in alphabet-
ical order (by author name).
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-2-36-S3.doc]
Additional file 4
Description of the biological content of the pathway. A description of the
signalling of the four pathways (Toll-like receptor, interferon, NF-?B and
apoptosis) depicted on the integrated pathway diagram is provided here.
The interconnectivity of these pathways and their significance in innate
immune signalling is also discussed in this section.
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-2-36-S4.doc]
Additional file 5
Interferon gamma regulated genes. A summary of the analysis of the 58
genes present on both the pathway map and in a transcriptional network
of differentially expressed genes following Ifng stimulation. A transcrip-
tional network of all differentially expressed genes (above 1.6 fold change)
was constructed and clustered using the graph-based clustering algorithm
MCL set at an inflation value of 2.2. This resulted in 26 different clusters,
which were then assigned a description of the co-expression pattern they
represent over the time course. The cluster numbers and the descriptions
of co-expression pattern are shown in this data sheet for the genes present
on the pathway diagram. 3 of these genes did not appear in any cluster.
Also included in this table is a summary of the gene expression changes
according to annova and Empirical Bayes calculations. RMA normalized
expression values are included for each gene across the time course as are
gene descriptions and GO (gene ontology) annotations for the 58 genes.
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-2-36-S5.xls]
Publish with BioMed Central and every
scientist can read your work free of charge
"BioMed Central will be the most significant development for
disseminating the results of biomedical research in our lifetime."
Sir Paul Nurse, Cancer Research UK
Your research papers will be:
available free of charge to the entire biomedical community
peer reviewed and published immediately upon acceptance
cited in PubMed and archived on PubMed Central
yours — you keep the copyright
Submit your manuscript here:
http://www.biomedcentral.com/info/publishing_adv.asp
BioMedcentral
BMC Systems Biology 2008, 2:36
http://www.biomedcentral.com/1752-0509/2/36
Page 15 of 15
(page number not for citation purposes)
inducible immediate-early gene encoding a short-lived glyc-
osylated protein. Oncogene 1993, 8(3):797-801.
33.
Navarro L, Mowen K, Rodems S, Weaver B, Reich N, Spector D,
David M: Cytomegalovirus activates interferon immediate-
early response gene expression and an interferon regulatory
factor 3-containing interferon-stimulated response element-
binding complex. Mol Cell Biol 1998, 18(7):3796-3802.
34.
Ohki R, Yamamoto K, Mano H, Lee RT, Ikeda U, Shimada K: Identi-
fication of mechanically induced genes in human monocytic
cells by DNA microarrays. J Hypertens 2002, 20(4):685-691.
35.
Pietzsch A, Buchler C, Aslanidis C, Schmitz G: Identification and
characterization of a novel monocyte/macrophage differen-
tiation-dependent gene that is responsive to lipopolysaccha-
ride, ceramide, and lysophosphatidylcholine. Biochem Biophys
Res Commun 1997, 235(1):4-9.
36.
Li H, Zhu H, Xu CJ, Yuan J: Cleavage of BID by caspase 8 medi-
ates the mitochondrial damage in the Fas pathway of apop-
tosis. Cell 1998, 94(4):491-501.
37.
Wen J, Ramadevi N, Nguyen D, Perkins C, Worthington E, Bhalla K:
Antileukemic drugs increase death receptor 5 levels and
enhance Apo-2L-induced apoptosis of human acute leuke-
mia cells. Blood 2000, 96(12):3900-3906.
38.
Yin XM: Signal transduction mediated by Bid, a pro-death
Bcl-2 family proteins, connects the death receptor and mito-
chondria apoptosis pathways. Cell Res 2000, 10(3):161-167.
39.
STRING: functional protein association networks [http://
string.embl.de/]
40.
Ingenuity Pathway Analysis Software [http://www.ingenu
ity.com]
|
18433497
|
TBK1 = ( External_Activator )
IL1R1 = ( External_Activator )
APAF1gene = ( TP53nucleus )
ILIB_IL1R1_MYD88_IRAK1_IRAK4 = ( ILIB_IL1R1 AND ( ( ( IRAK4 AND IRAK1 AND MYD88 ) ) ) )
TNFRSF10B = ( External_Activator )
RELAp65_NFKB1p50cytoplasm = ( NFKBIA_RELAp65_NFKB1p50 AND ( ( ( MAP3K7 OR RPS6KA5 OR IKBKB OR CHUK OR TBK1 OR PRKCZ ) ) ) )
BAX = ( Mitochondrial_Activator )
BCL2_BAX = ( BCL2 AND ( ( ( BAX ) ) ) )
TNF_BAG4_TNFRSF1A = ( TNF AND ( ( ( BAG4_TNFRSF1A ) ) ) )
EP300 = ( External_Activator )
STAT1gene = ( IRF1_IRF1nucleus )
PARP = ( External_Activator )
BCL3_NFKB2p52_NFKB2p52 = ( BCL3 AND ( ( ( NFKB2p52_NFKB2p52nucleus ) ) ) )
TLR9_TLR9 = ( TLR9 AND ( ( ( Viral_Bacterial_CpG ) ) ) )
IRF3_IRF3cytoplasm = ( IRF3 )
ISGF3cytoplasm = ( STAT1_STAT2 AND ( ( ( IRF9 ) ) ) )
LMNA = ( External_Activator )
TNFSF10_TNFRSF10A = ( TNFSF10 AND ( ( ( TNFRSF10A ) ) ) )
GAS2 = ( External_Activator )
IFNGR1 = ( External_Activator )
IRF1gene = ( STAT1_STAT1nucleus_p2 )
TNF_TNFRSF1B = ( TNF AND ( ( ( TNFRSF1B ) ) ) )
STAT1_STAT1_IRF9cytoplasm = ( STAT1 AND ( ( ( IRF9 ) ) ) )
TICAM1 = ( External_Activator )
BIRC4gene = ( RELAp65_NFBK1p50nucleus )
cLMNA = ( LMNA AND ( ( ( CASP6nucleus ) ) ) )
TOLLIP = ( External_Activator )
TNFSF10gene = ( IRF5_IRF5nucleus ) OR ( IRF1_IRF1nucleus )
STAT1_STAT1_IRF9nucleus = ( STAT1_STAT1_IRF9cytoplasm )
FASLGgene = ( TP53nucleus ) OR ( IRF1_IRF1nucleus )
TLR1_TLR2_TIRAP_MYD88_IRAK2_IRAK1_IRAK4_TRAF6 = ( TLR1_TLR2_TIRAP_MYD88_IRAK2_IRAK1_IRAK4 AND ( ( ( TRAF6 ) ) ) )
IRF5_IRF7nucleus = ( IRF5_IRF7cytoplasm )
Apoptosis = ( Inactive_DNA_Repair ) OR ( Cell_Shrinkage ) OR ( Inactivation_of_Protein_Synthesis ) OR ( DNA_Fragmentation )
HLA_Bgene = ( IRF8 )
RELAp65_NFBK1p50nucleus = ( RELAp65_NFKB1p50cytoplasm )
TLR3 = ( External_Activator )
IRAK1 = ( External_Activator )
BID = ( External_Activator )
IRF7_IRF7cytoplasm = ( IRF7 )
BCL2A1gene = ( RELAp65_NFBK1p50nucleus )
TNFSF13B_TNFRSF17_TRAF5 = ( TNFSF13B_TNFRSF17 AND ( ( ( TRAF5 ) ) ) )
PSMB9gene = ( IRF2 ) OR ( IRF1_IRF1nucleus )
IFNGR2 = ( External_Activator )
CXCL10gene = ( CBP AND ( ( ( IRF3_IRF7nucleus OR IRF3_IRF3nucleus ) ) ) ) OR ( ISGF3nucleus )
IRF5 = ( TLR7_MYD88_TRAF6_IRF5 ) OR ( Virus ) OR ( TLR9_MYD88_TRAF6_IRF5 )
TLR5 = ( External_Activator )
PDCD8_HSPA1A = ( PDCD8cytoplasm AND ( ( ( HSPA1A ) ) ) )
Cell_Shrinkage = ( cGAS2 ) OR ( cLMNA )
CHUK_CHUK = ( CHUK AND ( ( ( MAP3K14 ) ) ) )
JAK2 = ( External_Activator )
CBP = ( EP300 AND ( ( ( CREBBP ) ) ) )
CKII = ( IRF1_IRF1_Activator )
DIABLOcytoplasm = ( DIABLOmitochondria AND ( ( ( BAK1 OR BAX OR tBID ) ) ) )
IRF5_IRF5nucleus = ( IRF5_IRF5cytoplasm )
IRF7 = ( IKBKE_TBK1 )
CCL5gene = ( IRF1_IRF1nucleus )
MYD88 = ( External_Activator )
BAG4 = ( External_Activator ) OR ( TNF_BAG4_TNFRSF1A )
CYBBgene = ( IRF8 AND ( ( ( SPI1 ) ) ) )
ILIB_IL1R1_MYD88_IRAK1_IRAK4_TRAF6 = ( ILIB_IL1R1_MYD88_IRAK1_IRAK4 AND ( ( ( TRAF6 ) ) ) )
EIF2AK2_PRKRA = ( EIF2AK2cytoplasm AND ( ( ( PRKRA ) ) ) )
STAT1 = ( IFNGR )
IFNA_IFNA = ( IFNA )
BAK1gene = ( IRF5_IRF5nucleus )
ISGF3nucleus = ( ISGF3cytoplasm )
TLR4_TICAM1_TICAM2 = ( TLR4 AND ( ( ( TICAM2 AND TICAM1 ) ) ) )
SP100gene = ( IRF5_IRF5nucleus )
RELA_p65 = ( RELA_NFKB1_Activator )
TNFSF10_TNFSF10B_FADD = ( TNFSF10_TNFSF10B AND ( ( ( FADD ) ) ) )
IPARP = ( PARP AND ( ( ( CASP3nucleus OR CASP7nucleus ) ) ) )
TLR5_MYD88_IRAK1_IRAK4_TRAF6 = ( TLR5_MYD88_IRAK1_IRAK4 AND ( ( ( TRAF6 ) ) ) )
IFNB1_IFNB1 = ( IFNB1 )
IRF3_IRF7nucleus = ( IRF3_IRF7cytoplasm )
IRF1cytoplasm = ( PKC ) OR ( PKA ) OR ( CKII )
PTP = ( External_Activator )
TRAF2gene = ( RELAp65_NFBK1p50nucleus )
NFKB1_p50 = ( RELA_NFKB1_Activator )
IRAK4 = ( External_Activator )
BIRC3gene = ( RELAp65_NFBK1p50nucleus )
CASP9 = ( CASP6_Activator )
Inactivation_of_Protein_Synthesis = ( EIF2S1 )
FAF1 = ( External_Activator )
BCL3 = ( External_Activator )
TNF_TNFRSF1A = ( TNF_BAG4_TNFRSF1A )
CASP1 = ( ProCASP1 AND ( ( ( CASP4 ) ) ) )
CFLAR = ( External_Activator )
TLR2_TLR6_TOLLIP_MYD88_IRAK1_IRAK4 = ( TLR2_TLR6 AND ( ( ( TOLLIP AND IRAK4 AND IRAK1 AND MYD88 ) ) ) )
CHUK = ( External_Activator )
TRAF6 = ( External_Activator )
TLR2 = ( External_Activator )
TRAF1gene = ( RELAp65_NFBK1p50nucleus )
TLR3_TICAM1_TICAM2 = ( TLR3_TLR3 AND ( ( ( TICAM2 AND TICAM1 ) ) ) )
TNFRSF1A = ( External_Activator )
FASgene = ( RELAp65_NFBK1p50nucleus )
FAS = ( External_Activator )
TLR1_TLR2 = ( Triacyl_Lipopeptides )
FASLG_FAS = ( FASLG AND ( ( ( FAS ) ) ) )
TLR7 = ( External_Activator )
ProCASP8 = ( External_Activator )
IRF7_IRF7nucleus = ( IRF7_IRF7cytoplasm )
NFKB2p52_NFKB2p52nucleus = ( NFKB2p52_NFKB2p52cytoplasm )
CASP3cytoplasm = ( ProCASP3 AND ( ( ( Apoptosome OR CASP4 OR CASP8 ) ) ) )
TNF_TNFRSF1A_FADD_TRADD = ( TNF_TNFRSF1A AND ( ( ( TRADD AND FADD ) ) ) )
TLR9 = ( External_Activator )
BAD = ( Mitochondrial_Activator )
BAG4_TNFRSF1A = ( BAG4 AND ( ( ( TNFRSF1A ) ) ) )
CASP4 = ( ProCASP4 )
CASP7nucleus = ( CASP7cytoplasm )
BCL2A1 = ( RELAp65_NFBK1p50nucleus )
TLR3_TICAM1_TICAM2_TBK1 = ( TLR3_TICAM1_TICAM2 AND ( ( ( TBK1 ) ) ) )
Proteasome = ( External_Activator )
PDCD8mitochondria = ( Mitochondrial_Activator )
NFKB2p100_RELB_Ub = ( NFKB2p100_RELBcytoplasm AND ( ( ( Ub ) ) ) )
PTPN2 = ( External_Activator )
IKBKE_TBK1 = ( TLR4_TICAM1_TICAM2 AND ( ( ( TBK1 AND IKBKE ) ) ) )
CYCScytoplasm = ( CYCSmytochondria AND ( ( ( BAK1 OR BAX OR tBID ) ) ) )
IRF1_IRF1cytoplasm = ( IRF1cytoplasm )
BIRC4cytoplasm = NOT ( ( DIABLOcytoplasm ) OR ( HTRA2cytoplasm ) )
TLR1_TLR2_TIRAP_MYD88_IRAK2_IRAK1_IRAK4 = ( TLR1_TLR2 AND ( ( ( IRAK4 AND IRAK1 AND IRAK2 AND MYD88 AND TIRAP ) ) ) )
TLR5_MYD88_IRAK1_IRAK4 = ( TLR5_TLR5 AND ( ( ( IRAK4 AND IRAK1 AND MYD88 ) ) ) )
IRF1_IRF1nucleus = ( IRF1_IRF1cytoplasm )
PRKCZ = ( External_Activator )
NFKB2p52_RELBcytoplasm = ( NFKB2p100_RELB_Ub AND ( ( ( Proteasome ) ) ) )
RELB = ( External_Activator )
Diacyl_Lipopeptides = ( Bacteria )
SOD2gene = ( RELAp65_NFBK1p50nucleus )
NFKB2p100_NFKB2p100_Ub = ( NFKB2p100_NFKB2p100cytoplasm AND ( ( ( Ub ) ) ) )
RPS6KA5 = ( External_Activator )
IFNAR1 = ( External_Activator )
TLR3_TICAM1_TICAM2_TRAF6 = ( TLR3_TICAM1_TICAM2 AND ( ( ( TRAF6 ) ) ) )
PDCD8cytoplasm = ( tBID AND ( ( ( PDCD8mitochondria ) ) ) )
ENDOGcytoplasm = ( tBID AND ( ( ( ENDOGmitochondria ) ) ) )
FASLG_FAS_FADD_FAF1_DAXX = ( FASLG_FAS AND ( ( ( FAF1 AND FADD AND DAXX ) ) ) )
TLR3_TICAM1_TICAM2_RIPK1 = ( TLR3_TICAM1_TICAM2 AND ( ( ( RIPK1 ) ) ) )
NFKB2p52_RELBnucleus = ( NFKB2p52_RELBcytoplasm )
TNFRSF17 = ( External_Activator )
LPS = ( Bacteria )
ENDOGnucleus = ( ENDOGcytoplasm )
DNA_Fragmentation = ( IDFFA ) OR ( Fragmented_DNAnucleus )
HSPA1A = ( External_Activator )
Flagellin = ( Bacteria )
CXCL9gene = ( STAT1_STAT1nucleus_p2 )
TLR9_MYD88_TRAF6 = ( TLR9_MYD88_IRAK1_IRAK4_TRAF6 )
NFKB2p100 = ( External_Activator )
TLR3_TLR3 = ( dsRNA AND ( ( ( TLR3 ) ) ) )
ProCASP3 = ( CASP3_Activator )
IRF3_IRF3nucleus = ( IRF3_IRF3cytoplasm )
IRAK2 = ( External_Activator )
Inactive_DNA_Repair = ( IPARP )
TAP1gene = ( IRF1_IRF1nucleus ) OR ( IRF2 )
ProCASP7 = ( CASP7_Activator )
EIF2AK2cytoplasm = ( dsRNA )
ILIB_IL1R1 = ( ILIB AND ( ( ( IL1R1 ) ) ) )
BIRC2 = ( External_Activator )
STAT1_STAT1cytoplasm = ( STAT1 AND ( ( ( PRKCD ) ) ) )
STAT1_STAT2 = ( STAT1 AND ( ( ( STAT2 ) ) ) )
TRAF5 = ( External_Activator )
IRF3 = ( TLR3_TICAM1_TICAM2_TBK1 ) OR ( IKBKE_TBK1 )
IDFFA = ( DFFA AND ( ( ( CASP3nucleus ) ) ) )
TLR6 = ( External_Activator )
IFR5gene = ( STAT1_STAT1_IRF9nucleus ) OR ( TP53nucleus )
HIST2H4gene = ( IRF2 ) OR ( IRF1_IRF1nucleus )
IL12Bgene = ( IRF1_IRF1nucleus )
TNF_TNFRSF1B_TRAF2 = ( TNF_TNFRSF1B AND ( ( ( TRAF2cytoplasm ) ) ) )
CD40 = ( External_Activator )
IFNAR2 = ( External_Activator )
PRKCD = ( External_Activator )
TYK2 = ( External_Activator )
ENDOGmitochondria = ( Mitochondrial_Activator )
PKC = ( IRF1_IRF1_Activator )
TLR9_MYD88_IRAK1_IRAK4_TRAF6 = ( TLR9_MYD88_IRAK1_IRAK4 AND ( ( ( TRAF6 ) ) ) )
cGAS2 = ( GAS2 AND ( ( ( CASP3nucleus ) ) ) )
IER3gene = ( RELAp65_NFBK1p50nucleus )
FASLG_FAS_FADD_FAF1_DAXX_CFLAR = ( FASLG_FAS_FADD_FAF1_DAXX AND ( ( ( CFLAR ) ) ) )
CREBBP = ( External_Activator )
PLSCR1gene = ( IRF5_IRF5nucleus )
CD40_CD40LG = ( CD40 AND ( ( ( CD40LG ) ) ) )
TLR2_TLR6_TOLLIP_MYD88_IRAK1_IRAK4_TRAF6 = ( TLR2_TLR6_TOLLIP_MYD88_IRAK1_IRAK4 AND ( ( ( TRAF6 ) ) ) )
CXCL11gene = ( IRF5_IRF5nucleus )
TLR7_MYD88_TRAF6_IRF5 = ( TLR7_TLR7 AND ( ( ( TRAF6 AND MYD88 ) AND ( ( ( NOT IRF5 ) ) ) ) ) )
TLR2_TLR6 = ( TLR2 AND ( ( ( Diacyl_Lipopeptides AND TLR6 ) ) ) )
BCL2L1_BAD = ( BCL2L1mitochondria AND ( ( ( BAD ) ) ) )
NFKB2p100_RELBcytoplasm = ( NFKB2p100 AND ( ( ( CHUK_CHUK AND RELB ) ) ) )
TRADD = ( External_Activator )
ssRNA = ( Virus )
TNF_TNFRSF1A_FADD_TRADD_TRAF2_RIPK1 = ( TNF_TNFRSF1A_FADD_TRADD AND ( ( ( TRAF2cytoplasm AND RIPK1 ) ) ) )
TLR5_TLR5 = ( TLR5 AND ( ( ( Flagellin ) ) ) )
TP53nucleus = ( TP53cytoplasm )
Ub = ( External_Activator )
IKBKG_CHUK_IKBKB = ( TLR3_TICAM1_TICAM2_RIPK1_RIPK3 AND ( ( ( IKBKB AND CHUK AND IKBKG ) ) ) ) OR ( IKBKG AND ( ( ( IKBKB AND MAP3K7IP1_MAP3K7IP2_MAP3K7 AND CHUK ) ) ) )
MAP3K7IP2 = ( External_Activator )
DAXX = ( External_Activator )
dsRNA = ( Virus )
BCL2 = ( Mitochondrial_Activator )
CYCSmytochondria = ( Mitochondrial_Activator )
JAK1 = ( External_Activator )
TRAF3 = ( External_Activator )
IFNAR = ( IFNA AND ( ( ( IFNAR2 AND JAK1 AND TYK2 AND IFNAR1 ) ) ) ) OR ( IFNB1 AND ( ( ( IFNAR2 AND JAK1 AND TYK2 AND IFNAR1 ) ) ) )
TNF_IKBKG_Complex = ( TNF_TNFRSF1A_FADD_TRADD_TRAF2_RIPK1 AND ( ( ( IKBKB AND CHUK AND IKBKG ) ) ) )
CASP6cytoplasm = ( ProCASP6 AND ( ( ( CASP9 ) ) ) )
CASP10 = ( ProCASP10 AND ( ( ( TNF_TNFRSF1A_FADD_TRADD ) ) ) )
STAT2 = ( IFNAR )
MAP3K7IP1 = ( External_Activator )
IRF2gene = ( IRF1_IRF1nucleus )
PRKRA = ( External_Activator )
SOCS1 = ( External_Activator )
IFIT2gene = ( CBP AND ( ( ( IRF3_IRF7nucleus OR IRF3_IRF3nucleus ) ) ) ) OR ( ISGF3nucleus )
BBC3gene = ( TP53nucleus )
OAS1gene = ( IRF8 ) OR ( IRF1_IRF1nucleus ) OR ( ISGF3nucleus ) OR ( IRF5_IRF5nucleus )
IKBKE = ( External_Activator )
CIITAgene = ( IRF2 ) OR ( IRF1_IRF1nucleus )
MDM2gene = ( TP53nucleus )
NFKB2p100_NFKB2p100cytoplasm = ( NFKB2p100 AND ( ( ( IKBKG_CHUK_IKBKB ) ) ) )
RIPK3 = ( External_Activator )
Fragmented_DNAnucleus = ( DNA AND ( ( ( ENDOGnucleus OR PDCD8nucleus ) ) ) )
CASP3gene = ( IRF5_IRF5nucleus ) OR ( CASP3cytoplasm )
CFLARgene = ( RELAp65_NFBK1p50nucleus )
IL1Bgene = ( IRF8 AND ( ( ( SPI1 ) ) ) )
TNFSF10_TNFSF10B = ( TNFSF10 AND ( ( ( TNFRSF10B ) ) ) )
SPI1 = ( External_Activator )
IRF5_IRF7cytoplasm = ( IRF5 )
MAP3K7IP1_MAP3K7IP2_MAP3K7 = ( TLR1_TLR2_TIRAP_MYD88_IRAK2_IRAK1_IRAK4_TRAF6 AND ( ( ( MAP3K7 AND MAP3K7IP2 AND MAP3K7IP1 ) ) ) ) OR ( TLR3_TICAM1_TICAM2_TRAF6 AND ( ( ( MAP3K7 AND MAP3K7IP2 AND MAP3K7IP1 ) ) ) ) OR ( TLR7_MYD88_IRAK1_IRAK4_TRAF6 AND ( ( ( MAP3K7 AND MAP3K7IP2 AND MAP3K7IP1 ) ) ) ) OR ( TLR9_MYD88_IRAK1_IRAK4_TRAF6 AND ( ( ( MAP3K7 AND MAP3K7IP2 AND MAP3K7IP1 ) ) ) ) OR ( ILIB_IL1R1_MYD88_IRAK1_IRAK4_TRAF6 AND ( ( ( MAP3K7 AND MAP3K7IP2 AND MAP3K7IP1 ) ) ) ) OR ( TLR2_TLR6_TOLLIP_MYD88_IRAK1_IRAK4_TRAF6 AND ( ( ( MAP3K7 AND MAP3K7IP2 AND MAP3K7IP1 ) ) ) ) OR ( TLR5_MYD88_IRAK1_IRAK4_TRAF6 AND ( ( ( MAP3K7 AND MAP3K7IP2 AND MAP3K7IP1 ) ) ) )
RIPK1 = ( External_Activator )
RIPK1gene = ( IRF5_IRF5nucleus )
IRF3_IRF5nucleus = ( IRF3_IRF5cytoplasm )
Fragmented_DNAcytoplasm = ( DNA_Fragmentation )
EIF2AK2gene = ( IRF1_IRF1nucleus ) OR ( ISGF3nucleus )
HTRA2mitochondria = ( Mitochondrial_Activator )
DFFA = ( External_Activator )
BAK1 = ( Mitochondrial_Activator )
ProCASP10 = ( External_Activator )
CASP7cytoplasm = ( ProCASP7 AND ( ( ( Apoptosome OR CASP8 OR CASP10 ) ) ) )
IKBKB = ( External_Activator )
IKBKG = ( External_Activator )
CASP2 = ( ProCASP2 AND ( ( ( Fragmented_DNAcytoplasm ) ) ) )
GBP1gene = ( IRF2 ) OR ( IRF1_IRF1nucleus )
TNFRSF1B = ( External_Activator )
IRF3_IRF7cytoplasm = ( IRF3 AND ( ( ( IRF7 ) ) ) )
MAP3K14 = ( CD40_CD40LG_TRAF3 ) OR ( TNFSF13B_TNFRSF17_TRAF5 ) OR ( TNF_TNFRSF1B_TRAF2 )
FADD = ( External_Activator )
Triacyl_Lipopeptides = ( Bacteria )
Apoptosome = ( APAF1_CYCS AND ( ( ( CASP9 ) ) ) )
IFIT1gene = ( IRF5_IRF5nucleus )
tBID = ( BID AND ( ( ( CASP8 OR CASP2 ) ) ) )
DNA = ( External_Activator )
IRF2 = ( IRF2_Activator )
ProCASP4 = ( External_Activator )
IFNGR = ( IFNG AND ( ( ( IFNGR2 AND IFNGR1 AND JAK2 ) ) ) )
TLR3_TICAM1_TICAM2_RIPK1_RIPK3 = ( TLR3_TICAM1_TICAM2_RIPK1 AND ( ( ( RIPK3 ) ) ) )
CD40_CD40LG_TRAF3 = ( CD40_CD40LG AND ( ( ( TRAF3 ) ) ) )
TP53cytoplasm = ( EIF2AK2_PRKRA )
ICAM1gene = ( STAT1_STAT1nucleus_p2 )
NFKB2p52_NFKB2p52cytoplasm = ( NFKB2p100_NFKB2p100_Ub AND ( ( ( Proteasome ) ) ) )
TLR9_MYD88_TRAF6_IRF5 = ( TLR9_TLR9 AND ( ( ( TRAF6 AND MYD88 ) AND ( ( ( NOT IRF5 ) ) ) ) ) )
PDCD8nucleus = ( PDCD8cytoplasm )
G1P2gene = ( CBP AND ( ( ( IRF3_IRF7nucleus OR IRF3_IRF3nucleus ) ) ) ) OR ( IRF1_IRF1nucleus AND ( ( ( IRF8 AND SPI1 AND IRF4 ) ) ) ) OR ( ISGF3nucleus ) OR ( IRF2 AND ( ( ( IRF8 AND SPI1 AND IRF4 ) ) ) )
TNFSF13B_TNFRSF17 = ( TNFSF13B AND ( ( ( TNFRSF17 ) ) ) )
ATF2 = ( External_Activator )
Viral_Bacterial_CpG = ( Virus ) OR ( Bacteria )
CASP3nucleus = ( CASP3cytoplasm )
TLR7_MYD88_IRAK1_IRAK4_TRAF6 = ( TLR7_MYD88_IRAK1_IRAK4 AND ( ( ( TRAF6 ) ) ) )
IRF4 = ( External_Activator )
ProCASP6 = ( CASP6_Activator )
TLR9_MYD88_IRAK1_IRAK4 = ( TLR9_TLR9 AND ( ( ( IRAK4 AND IRAK1 AND MYD88 ) ) ) )
TNFRSF10Bgene = ( TP53nucleus )
BCL2L1gene = ( RELAp65_NFBK1p50nucleus )
HTRA2cytoplasm = ( tBID AND ( ( ( HTRA2mitochondria ) ) ) )
EIF2S1 = ( EIF2AK2gene )
PKA = ( IRF1_IRF1_Activator )
TNFSF10_TNFRSF10A_FADD = ( TNFSF10_TNFRSF10A AND ( ( ( FADD ) ) ) )
TLR7_MYD88_IRAK1_IRAK4 = ( TLR7_TLR7 AND ( ( ( IRAK4 AND IRAK1 AND MYD88 ) ) ) )
G1P3gene = ( IRF8 )
CASP8 = ( ProCASP8 AND ( ( ( TNFSF10_TNFRSF10A_FADD OR FASLG_FAS_FADD_FAF1_DAXX OR FASLG_FAS_FADD_FAF1_DAXX_CFLAR OR TNFSF10_TNFSF10B_FADD ) ) ) )
TRAF2cytoplasm = ( External_Activator )
STAT1_STAT1nucleus_p2 = ( STAT1_STAT1nucleus_p1 )
STAT1_STAT1nucleus_p1 = ( STAT1_STAT1cytoplasm )
NFKBIA = ( NFKBIA_RELAp65_NFKB1p50 ) OR ( RELA_NFKB1_Activator )
IRF9 = ( External_Activator )
IRF8 = ( IRF2 ) OR ( IRF1_IRF1nucleus )
TLR7_TLR7 = ( TLR7 AND ( ( ( ssRNA ) ) ) )
APAF1_CYCS = ( APAF1 AND ( ( ( CYCScytoplasm ) ) ) )
PMAIPgene = ( IRF5_IRF5nucleus )
ProCASP2 = ( External_Activator )
TICAM2 = ( External_Activator )
SOCS3 = ( External_Activator )
IFNAgene = ( STAT1_STAT1_IRF9nucleus ) OR ( IRF3_IRF3nucleus ) OR ( IRF1_IRF1nucleus ) OR ( ISGF3nucleus ) OR ( IRF5_IRF5nucleus ) OR ( IRF7_IRF7nucleus ) OR ( IRF3_IRF5nucleus ) OR ( IRF3_IRF7nucleus )
TLR4 = ( LPS )
TIRAP = ( External_Activator )
IRF5_IRF5cytoplasm = ( IRF5 )
ProCASP1 = ( External_Activator )
IRF3_IRF5cytoplasm = ( IRF5 )
DIABLOmitochondria = ( Mitochondrial_Activator )
CASP6nucleus = ( CASP6cytoplasm )
NOS2Agene = ( External_Activator )
PRKRAgene = ( IRF5_IRF5nucleus )
TNFRSF10A = ( External_Activator )
APAF1 = ( External_Activator )
IL15gene = ( IRF1_IRF1nucleus )
IFNB1gene = ( STAT1_STAT1nucleus_p2 ) OR ( ISGF3nucleus ) OR ( IRF1_IRF1nucleus ) OR ( IRF3_IRF3nucleus ) OR ( IRF5_IRF5nucleus ) OR ( RELAp65_NFBK1p50nucleus AND ( ( ( CBP AND IRF3_IRF7nucleus AND IRF3_IRF3nucleus AND ATF2 ) ) ) )
BCL2L1mitochondria = ( Mitochondrial_Activator )
MAP3K7 = ( External_Activator )
NFKBIA_RELAp65_NFKB1p50 = ( TNF_IKBKG_Complex AND ( ( ( NFKB1_p50 AND RELA_p65 AND NFKBIA ) ) ) ) OR ( IKBKG_CHUK_IKBKB AND ( ( ( NFKB1_p50 AND RELA_p65 AND NFKBIA ) ) ) )
|
Network model of survival signaling in large granular
lymphocyte leukemia
Ranran Zhang†, Mithun Vinod Shah†, Jun Yang†, Susan B. Nyland†, Xin Liu†, Jong K. Yun‡, Re´ka Albert§¶,
and Thomas P. Loughran, Jr.†
†Penn State Hershey Cancer Institute and ‡Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033;
and §Department of Physics, The Pennsylvania State University, University Park, PA 16802
Edited by Wayne M. Yokoyama, Washington University School of Medicine, St. Louis, MO, and approved September 4, 2008 (received for review
July 5, 2008)
T cell large granular lymphocyte (T-LGL) leukemia features a clonal
expansion of antigen-primed, competent, cytotoxic T lymphocytes
(CTL). To systematically understand signaling components that
determine the survival of CTL in T-LGL leukemia, we constructed a
T-LGL survival signaling network by integrating the signaling
pathways involved in normal CTL activation and the known de-
regulations of survival signaling in leukemic T-LGL. This network
was subsequently translated into a predictive, discrete, dynamic
model. Our model suggests that the persistence of IL-15 and PDGF
is sufficient to reproduce all known deregulations in leukemic
T-LGL. This finding leads to the following predictions: (i) Inhibiting
PDGF signaling induces apoptosis in leukemic T-LGL. (ii) Sphin-
gosine kinase 1 and NFB are essential for the long-term survival
of CTL in T-LGL leukemia. (iii) NFB functions downstream of PI3K
and prevents apoptosis through maintaining the expression of
myeloid cell leukemia sequence 1. (iv) T box expressed in T cells
(T-bet) should be constitutively activated concurrently with NFB
activation to reproduce the leukemic T-LGL phenotype. We vali-
dated these predictions experimentally. Our study provides a
model describing the signaling network involved in maintaining
the long-term survival of competent CTL in humans. The model will
be useful in identifying potential therapeutic targets for T-LGL
leukemia and generating long-term competent CTL necessary for
tumor and cancer vaccine development.
discrete dynamic model nuclear factor kappa-B
signal transduction network T box expressed in T cells
T cell large granular lymphocyte leukemia
C
ytotoxic T lymphocyte (CTL) activation normally involves
an initial expansion of antigen-specific CTL clones and their
acquisition of cytotoxic activity. Subsequently, the activated CTL
population undergoes activation-induced cell death (AICD),
resulting in final stabilization of a small antigen-experienced
CTL population (1). This process requires a delicate balance
between proliferation, survival, and apoptosis. T cell large
granular lymphocyte (T-LGL) leukemia is characterized by
abnormal clonal expansion of antigen-primed mature CTL that
successfully escaped AICD and remain long-term competent
(2). Similar to normal activated CTL, leukemic T-LGL exhibit
activation of multiple survival signaling pathways (3–5). How-
ever, unlike normal activated CTL, leukemic T-LGL are not
sensitive to Fas-induced apoptosis (6), a process essential for
AICD (7). Recent molecular profiling data suggest that normal
CTL activation and AICD are uncoupled in leukemic T-LGL
(8), providing a unique opportunity to decipher the key medi-
ators of CTL activation and AICD in humans.
Network modeling has been increasingly used to better un-
derstand complex and interactive biological systems (9, 10).
Experimentally obtained signaling pathway information can be
translated into a graph (network) by representing proteins,
transcripts, and small molecules as network nodes and denoting
the interactions between nodes as edges (9). The direction of
edges follows the direction of the mass or information flow, from
the upstream (source) node to the downstream (product or
target) node. In addition, the edges are characterized by signs,
where a positive sign indicates activation, and a negative sign
indicates inhibition. Discrete dynamic modeling is widely used in
modeling regulatory and signaling networks because of its
straightforwardness, robustness, and compatibility with qualita-
tive data (9–11). The simplest discrete models, called Boolean
models, assume two possible states for each node in the network:
ON (above threshold) and OFF (below threshold). The biolog-
ical functions by which upstream regulators act on a downstream
node can be readily translated into logical statements by using
Boolean operators.
In this study, we aimed to systematically understand the
long-term survival of competent CTL in T-LGL leukemia by
constructing a T-LGL survival signaling network and a Boolean
model of the network’s dynamics. We found the constitutive
presence of IL-15 and PDGF to be sufficient to reproduce all of
the other signaling abnormalities. In addition, we studied the
predicted key mediators of long-term CTL survival and their
related signaling pathways.
Results
Constructing the T-LGL Survival Signaling Network. We performed
an extensive literature search and constructed the T-LGL sur-
vival signaling network (shown in Fig. 1) by adapting and
simplifying a network describing the normal CTL activation–
AICD process. The detailed method of network construction is
described in supporting information (SI) Text. The information
used to construct the network, summarized by giving the source
node, target node, two qualifiers of the relationship, and refer-
ences, is given in Table S1. The nomenclature of all of the nodes
of the network before and after simplification is provided in
Tables S2 and S3. The T-LGL survival signaling network incor-
porates the most unique interactions through which all known
deregulations in leukemic T-LGL are connected, in the context
of normal CTL activation and AICD signaling. Proteins, mRNAs,
and small molecules (such as lipids) were represented as nodes.
‘‘Cytoskeleton signaling’’, ‘‘Proliferation’’ and ‘‘Apoptosis’’ were
also included as nodes to summarize the biological effects of a
group of components in the signaling pathways and serve as the
Author contributions: R.A. and T.P.L. designed research; R.Z., M.V.S., J.Y., and S.B.N.
performed research; X.L. and J.K.Y. contributed new reagents/analytic tools; R.Z., M.V.S.,
J.Y., S.B.N., X.L., J.K.Y., R.A., and T.P.L. analyzed data; and R.Z., R.A., and T.P.L. wrote the
paper.
The authors declare no conflict of interest.
This article is a PNAS Direct Submission.
¶To whom correspondence may be addressed at: Department of Physics, 122 Davey Labo-
ratory, The Pennsylvania State University, University Park, PA 16802. E-mail: ralbert@
phys.psu.edu.
To whom correspondence may be addressed at: Penn State Hershey Cancer Institute, 500
University Drive, Hershey, PA 17033. E-mail: tloughran@psu.edu.
This article contains supporting information online at www.pnas.org/cgi/content/full/
0806447105/DCSupplemental.
© 2008 by The National Academy of Sciences of the USA
16308–16313
PNAS
October 21, 2008
vol. 105
no. 42
www.pnas.orgcgidoi10.1073pnas.0806447105
indicators of cell fate. Because of the unknown etiology of
T-LGL leukemia (2), we used ‘‘Stimuli’’ as a node to indicate
antigen stimulation (12). This network contains 58 nodes and 123
edges. The biological description of the T-LGL survival signaling
network is given in SI Text.
Translating the T-LGL Survival Signaling Network into a Predictive,
Discrete, Dynamic Model. To understand the dynamics of signaling
abnormalities in T-LGL leukemia, we translated the T-LGL
survival signaling network into a Boolean model. Each network
node was described by one of two possible states: ON or OFF.
The ON state means the production of a small molecule, the
production and translation of a transcript, or the activation of a
protein/process whereas the OFF state means the absence of a
small molecule or transcript or the inhibition of a protein/
process. The regulation of each component in the network was
described by using the Boolean logical operators OR, AND, and
NOT (see Table S4). OR represents the combined effect of
independent upstream regulators on a downstream node
whereas AND indicates the conditional dependency of upstream
regulators to achieve a downstream effect. NOT represents the
effect of inhibitory regulators and can be combined with acti-
vating regulations by using either OR or AND. The rules were
derived from the regulatory relationships reflected in the net-
work and from the literature. The detailed justification of the
logical rules for all nodes in the network is provided in SI Text.
As in the biological system, there is a time lag between the state
change of the regulators and the state change of the targets. The
kinetics of signal propagation is rarely known from experiments.
Thus, we used an asynchronous updating algorithm (10, 11) that
samples differences in the speed of signal propagation. The
detailed algorithm is described in SI Text.
To reproduce how a population of cells responds to the same
signal and to simulate cell-to-cell variability, we performed
multiple simulations with the same initial conditions but differ-
ent updating orders (i.e., different timing). The model was
allowed to update for multiple rounds until the node Apoptosis
became ON in all simulations (recapitulating the death of all
CTL) or stabilized in the OFF state in a fraction of simulations
(recapitulating the stabilization of the long-term surviving CTL
population). The state of Stimuli was set to ON at the beginning
of every simulation, recapitulating the activation of CTL by
antigen. The states of the other nodes were set according to their
states in resting T cells, as described in the SI Text. At the end
of the simulation, if the state of a node stabilized at ON even
though it was OFF at the beginning of the simulation, we
consider it as constitutively active. If the state of a node
stabilized at OFF even though it was in the ON state at the
beginning of the simulation, or it was experimentally shown to
be active after normal CTL activation, we consider it as down-
regulated/inhibited. During simulations, the state of a node can
be fixed to reproduce signaling perturbations.
Constitutive Presence of IL-15 and PDGF Is Predicted to Be Sufficient
to Induce All of the Known Signaling Abnormalities in Leukemic T-LGL.
Zambello et al. (13) has demonstrated the presence of mem-
brane-bound IL-15 on leukemic LGL, suggesting a role of IL-15
in the pathogenesis of this disease. In the course of studying
constitutive cytokine production in LGL leukemia (14), we used
a protein array as an experimental method. Using this array, we
had found high levels of PDGF in LGL leukemia sera (unpub-
lished observation). PDGF exists in the form of homodimers or
heterodimers of two polypeptides: PDGF-A and PDGF-B (15).
In the current study, we examined the level of PDGF-BB level
in the sera of 22 T-LGL leukemia patients and 39 healthy donors
and found that PDGF-BB was significantly higher in patient
serum compared with normal (P 0.005) (Fig. 2A). We
subsequently incorporated this deregulation into the network
model.
To investigate signaling abnormalities underlying long-term
survival of leukemic T-LGL, we first tested whether our model
could reproduce the uncoupling of CTL activation and AICD by
using all known deregulations (summarized in Table S5). We did
not observe the activation of the node Apoptosis in any simu-
lation. Second, we probed whether all of the deregulations have
to be individually initiated or whether a subset of them can cause
the others. The effect of a single signaling perturbation can be
identified by keeping the state of the corresponding node
according to its deregulation and tracking the states of other
nodes until a stable (time-independent) state is obtained. IL-15,
PDGF, and Stimuli are three nodes that have been suggested to
be abnormal in T-LGL leukemia without known upstream
regulators in the T-LGL survival-signaling network. To recapit-
ulate the effect of their deregulations without masking the effect
of the perturbation tested, the states of IL-15, PDGF, and
Stimuli were randomly set at ON or OFF at every round of
Fig. 1.
The T-LGL survival signaling network. Node and edge color represents the current knowledge of the signaling abnormalities in T-LGL leukemia.
Up-regulated or constitutively active nodes are in red, down-regulated or inhibited nodes are in green, nodes that have been suggested to be deregulated (either
up-regulation or down-regulation) are in blue, and the states of white nodes are unknown or unchanged compared with normal. Blue edge indicates activation
and red edge indicates inhibition. The shape of the nodes indicates the cellular location: rectangular indicates intracellular components, ellipse indicates
extracellular components, and diamond indicates receptors. Conceptual nodes (Stimuli, Cytoskeleton signaling, Proliferation, and Apoptosis) are labeled orange.
The full names of the node labels are provided in Table S3.
Zhang et al.
PNAS
October 21, 2008
vol. 105
no. 42
16309
MEDICAL SCIENCES
updating, i.e., in a random state, except when probing for the
effect of their own deregulations.
Through this analysis, we discovered a hidden hierarchy
among the known deregulations in leukemic T-LGL, with up-
stream deregulations as the potential cause of downstream
deregulations. We present these regulatory relationships as a
hierarchical network in Fig. 2B. Details of the hierarchy analysis
are provided in SI Text. Surprisingly, we found that keeping the
state of IL-15 at ON was sufficient to reproduce all known
deregulations in leukemic T-LGL when setting a random state
for PDGF and Stimuli (Fig. S1). To understand the effect of
PDGF and Stimuli individually upon the constant presence of
IL-15, we probed all of the possible states of PDGF and Stimuli.
We determined that the presence of PDGF is needed for the
long-term survival of leukemic T-LGL. In contrast, the consti-
tutive presence of Stimuli is not required after its initial activa-
tion (Fig. 2C). We concluded that based on the available
signaling information regarding T cell activation and AICD, the
minimal condition required for our model to reproduce all
known signaling abnormalities in T-LGL leukemia (i.e., a T-
LGL-like state) is IL-15 constantly ON, PDGF intermittently
ON, and Stimuli ON in the initial condition.
To test the unexpected prediction that PDGF signaling,
together with IL-15, maintains leukemic T-LGL survival, we
inhibited PDGF receptor by using its specific inhibitor AG 1296.
As shown in Fig. 2D, AG 1296 induced apoptosis specifically in
T-LGL leukemia peripheral blood mononuclear cells (PBMCs)
but not in normal PBMCs (n 3–4, P 0.03).
Sphingosine kinase 1 (SPHK1) Is Important for the Survival of Leuke-
mic T-LGL. The next question we asked was: Can we identify the
key mediators that determine the uncoupling of CTL activation
and AICD in T-LGL leukemia? In our experimental system, the
indication that a protein (small molecule or complex) is a key
mediator in the finding that altering its amount or function can
induce apoptosis in leukemic T-LGL. Accordingly, a corre-
sponding network node is a key mediator if its state stabilizes
once a T-LGL-like state is achieved, and altering its state
increases the frequency of the ON state of Apoptosis. The
detailed method to identify key mediators is provided in SI Text.
As summarized in Table S6, experiments already suggested nine
key mediators. We first tested whether our model could identify
the corresponding nodes as key mediators. A rapid increase of
apoptosis frequency was observed after resetting and maintain-
ing the states of all of the known key mediators individually to
their opposite states (from ON to OFF or from OFF to ON) after
reproducing a T-LGL-like state. Examples of simulation results
are provided in Fig. S2.
Based on this result, we systematically simulated the effect of
individually altering the states of all nodes that stabilize when a
T-LGL-like state is achieved. A list of these nodes is provided in
Table S7. In addition to PDGF and its receptor, the model
predicted seven additional key mediators: SPHK1, NFB, S1P,
SOCS, GAP, BID, and IL2RB, all exhibiting a similar dynamics
of inducing apoptosis in the model. Fig. 3A shows the effect of
inhibiting SPHK1 as an example. Recently, we found that the
sphingolipid signaling is deregulated in leukemic LGL (8). Thus,
we tested the effect of SPHK1 inhibition on leukemic T-LGL
survival experimentally by using its chemical inhibitors, SPHK1
inhibitor-I and -II (SKI-I and SKI-II) (16, 17). As shown in Figs.
3 B and C, both SKI-I and SKI-II significantly induced apoptosis
in T-LGL leukemia PBMCs in a dose-dependent manner but not
in normal PBMCs (n 4–6, P 0.03).
NFB Maintains the Survival of Leukemic T-LGL Through STAT3-
Independent Regulation of myeloid cell leukemia sequence 1 (Mcl-1).
Our model predicts that NFB is constitutively active and is a key
mediator of the survival of leukemic T-LGL (Fig. 4A). Consid-
ering its importance in regulating T cell proliferation, cytotox-
icity, and survival (18), we studied the activity and function of
NFB in T-LGL leukemia. As shown in Fig. 4B, nuclear extracts
of normal PBMCs rarely exhibited NFB activity as assessed by
EMSA whereas NFB activity was detected in most of the
nuclear extracts of T-LGL leukemia PBMCs.
Next, we examined the effect of inhibiting NFB by using its
specific inhibitor BAY 11–7082 (19, 20). As shown in Fig. 4C,
BAY 11–7082 inhibited the constitutive activity of NFB in
T-LGL leukemia PBMCs in a dose-dependent manner as as-
Fig. 2.
The Boolean model of the T-LGL survival signaling network predicts
that constitutive presence of IL-15, and PDGF is sufficient to induce all of the
known deregulations in T-LGL leukemia. (A) PDGF-BB is elevated in T-LGL
leukemia patient sera compared with normal. Serum level of PDGF-BB from 39
healthy donors (gray triangles) and 22 T-LGL leukemia patients (white dia-
monds) was assessed by using ELISA. The figure shows a 1.4-fold increase of
mean serum level of PDGF-BB (black bar) in T-LGL leukemia patients compared
with normal (*, P 0.005). (B) Hierarchy among known signaling deregula-
tions in T-LGL leukemia. Color code for nodes and edges is the same as in Fig.
1. (C) The effects of IL15, PDGF, and Stimuli on the frequency of apoptosis
during simulation. Keeping PDGF ON does not prevent the onset of apoptosis
(white triangles). While keeping IL-15 ON, keeping PDGF OFF from the first
round of updating delays but cannot prevent the onset of apoptosis (white
squares). Setting Stimuli ON at the beginning of the simulation and then
keeping it OFF (‘‘ONCE’’) does not alter the inhibition of apoptosis upon
keeping IL-15 ON in the presence of PDGF (white circles). Results were ob-
tained from 400 simulations of each initial condition. (D) 10 M AG 1296
specifically induced apoptosis in T-LGL leukemia PBMCs (white circles, n 4)
after 24 h but not in normal PBMCs (gray circles, n 3, *, P 0.03). Each circle
represents data from one patient or healthy donor. The markers (black bars)
indicate the mean apoptosis percentage.
16310
www.pnas.orgcgidoi10.1073pnas.0806447105
Zhang et al.
sessed by EMSA. BAY 11–7082 (1 M) significantly induced
apoptosis in T-LGL leukemia PBMCs but not in normal PBMCs
(n 6, P 0.009) (Fig. 4D).
Based on the Boolean logical rule for NFB (see Table S4),
it can be activated by PI3K or tumor progression locus 2
(TPL2)/cancer Osaka thyroid. During simulations, however, we
noticed that the ON state of NFB was only determined by the
constitutive activity of PI3K because resetting the state of PI3K
from ON to OFF was necessary and sufficient to induce the rapid
inhibition of NFB whereas altering the state of TPL2 did not
affect NFB activity (Fig. 5A). We then subjected the T-LGL
leukemia PBMCs to PI3K specific inhibitor LY 294002 and
tested the effect on NFB activity. LY 294002 (25 M) signif-
icantly inhibited the activity of NFB as assessed by EMSA (Fig.
5B) and induced apoptosis in T-LGL leukemia PBMCs (data not
shown) as reported (4). BAY 11–7082 did not inhibit the activity
(phosphorylation) of v-akt murine thymoma viral oncogen ho-
molog (AKT), an immediate downstream target of PI3K in T-LGL
leukemia PBMCs, as assessed by Western blot assay (Fig. 5C).
We found that for simulations in which we reset the state of
NFB from ON to OFF, the onset of apoptosis correlated tightly
with the rapid down-regulation of Mcl-1 (Fig. 4A). It has been
shown that STAT3 transcriptionally regulates Mcl-1 in leukemic
T-LGL (3). Interestingly, our model predicted that the initial
Mcl-1 down-regulation after NFB inhibition was independent
of STAT3 activity (Fig. 4A). Our experimental results confirmed
this prediction. As shown in Fig. 5C, the amount of Mcl-1 after
BAY 11–7082 treatment did indeed decrease correspondingly to
the decreased NFB activity whereas STAT3 activity remained
unchanged after NFB inhibition, as assessed by EMSA, even at
the highest dose of BAY 11–7082.
T box expressed in T cells (T-bet) Is Constitutively Active in Leukemic
T-LGL. It has been shown that leukemic T-LGL are incapable of
producing IL-2 even upon in vitro stimulation (21). It has also
been shown that NFB promotes IL2 transcription (18). In our
model, this apparent conflict can be resolved if T-bet is consti-
Fig. 3.
SPHK1 is a key mediator for the survival of leukemic T-LGL. (A) The
effect of SPHK1 inhibition on Apoptosis frequency in the model. The state of
SPHK1 was reset to OFF after 15 rounds of updating (white squares), or left
unchanged (white diamonds) after a T-LGL-like state was achieved. A rapid
increase of apoptosis was observed after SPHK1 inhibition (200 simulations).
(B) 20 M and 40 M SKI-I selectively induced apoptosis in T-LGL leukemia
PBMCs (n 6) after 48 h but not in normal PBMCs (n 5, *, P 0.03 and **,
P 0.01). Each circle represents data from one patient or healthy donor. The
markers (black bars) indicate the mean apoptosis percentage. (C) 5 M and 10
M SKI-II selectively induced apoptosis in T-LGL leukemia PBMCs after 48 h
(white circles, n 5) but not in normal PBMCs (gray circles, n 4, *, P 0.02,
and **, P 0.001). Each circle represents data from one patient or healthy
donor. The markers (black bars) indicate the mean apoptosis percentage.
Fig. 4.
NFB is constitutively active in T-LGL leukemia and mediates survival
of leukemic T-LGL. (A) Model prediction of the effects of NFB inhibition (200
simulations). The state of NFB was reset from ON to OFF after 15 rounds of
updating, while keeping IL-15 and PDGF ON. Apoptosis (black squares) was
rapidly induced after inhibiting NFB (black diamonds). The induction of
apoptosis was tightly coupled with the down-regulation of Mcl-1 (). In
contrast, the state of STAT3 (white triangles) remained unchanged until the
simulation was terminated. (B) NFB activity in nuclear extracts of PBMCs from
healthy donors and T-LGL leukemia patients. EMSA results are representative
of 16 healthy donors and 8 T-LGL leukemia patients tested. White space has
been inserted to indicate realigned gel lanes. (C) BAY 11–7082 inhibits NFB
activity in T-LGL leukemia PBMCs. T-LGL leukemia PBMCs were treated with
vehicle DMSO or 1 M, 2 M, or 5 M BAY 11–7082 for 3 h, and the activity of
NFB was assessed by EMSA. Result is representative of experiments in three
patients. (D) Compared with normal PBMCs (black circles, n 6), 1 M BAY
11–7082 selectively induced apoptosis in T-LGL leukemia PBMCs (white circles,
n 6) after 12h treatment (*, P 0.02). Each circle represents data from one
patient or healthy donor. The markers (black bars) indicate the mean of each
sample group.
0
0.2
0.4
0.6
0.8
1
0
5
10 15 20 25
Rounds of updating
Frequency of node
activation
Re-set PI3K from ON to OFF
0
0.2
0.4
0.6
0.8
1
0
5
10 15 20 25
Rounds of updating
Re-set TPL2 from ON to OFF
Frequency of node
activation
0 25
NFκB Binding
Free Probe
LY 294002 (µM)
p-AKT (Ser473)
AKT
IB
EMSA
GAPDH
GAPDH
BAY 11-7082 (µM)
0 1 2 5
p-AKT (Ser473)
AKT
Mcl-1
STAT3 binding
Free probe
IB
EMSA
A
B
C
PI3K
NFκB
X
TPL2
PI3K
NFκB
X
TPL2
Fig. 5.
NFB-mediated survival pathway in T-LGL leukemia involves PI3K and
Mcl-1. (A) Analysis of the potential cause(s) of the constitutive activation of
NFB. As in Table S4, the Boolean logical rule governing the state of NFB is
‘‘NFKB* [(TPL2 or PI3K) or (FLIP and TRADD and IAP)] and not Apoptosis’’.
When a T-LGL-like state is achieved, the state of TRADD stabilizes at OFF (see
Table S7). Thus, the node that activates NFB can only be TPL2 or PI3K, which
are known to be constitutively active in T-LGL leukemia (see Table S5). Rapid
inhibition of NFB (white squares) was observed after inhibiting PI3K (X) but
not after inhibiting TPL2 (gray triangles) (200 simulations). (B) PI3K inhibition
induced NFB inhibition in T-LGL leukemia. T-LGL leukemia PBMCs were
treated with vehicle DMSO or 25 M LY 294002 for 4 h. The amount of total
and phospho-AKT was assessed by Western blot assay; NFB activity was
assessed by EMSA. Result is representative of experiments in three patients. (C)
NFB inhibition down-regulates Mcl-1 but does not influence STAT3 activity
and the PI3K pathway. T-LGL leukemia PBMCs were treated with vehicle
DMSO or 1 M, 2 M or 5 M BAY 11–7082 for 3 h, and the amount of Mcl-1,
total- and phospho-AKT was assessed by Western blot assay. STAT3 activity
was assessed by EMSA. Result is representative of experiments in three pa-
tients. GAPDH was used as a loading control for all of the Western blot assays.
Zhang et al.
PNAS
October 21, 2008
vol. 105
no. 42
16311
MEDICAL SCIENCES
tutively active. As shown in Fig. 6A, when a T-LGL-like state was
achieved, the state of T-bet stabilized at ON, and there was a
rapid increase of IL-2 production when the state of T-bet was
reset to OFF. In agreement with the model’s prediction, T-bet
was significantly elevated in T-LGL leukemia PBMCs compared
with normal PBMCs at both mRNA and protein levels, as
assessed by real-time PCR and Western blot assay (Figs. 6 B and
C). In addition, T-LGL leukemia PBMCs exhibited high T-bet
activity as assessed by EMSA whereas the T-bet activity was
almost undetectable in normal PBMCs (Fig. 6D).
Discussion
The long-term survival of competent CTL in T-LGL leukemia
offers a unique opportunity to reveal the key mediators of CTL
activation and AICD in humans. In this study, we curated the
signaling pathways involved in normal CTL activation, AICD,
and the deregulations in leukemic T-LGL and compiled them
into a T-LGL survival signaling network. By formulating a
Boolean dynamic model of this network, we were able to identify
the potential causes and key regulators of this abnormal survival.
Signaling pathways are complex and dynamic. However, ex-
perimental results are usually focused on limited interactions of
the components in one pathway and neglecting the broader
effects in the same or other pathways. Integrating existing
pathway information to infer cross-talk among pathways is
desirable, especially for studies in T-LGL leukemia where ex-
perimental data are limited. Using network theory as a tool, we
identified the most unique interactions among known deregu-
lated components in leukemic T-LGL in the context of CTL
activation and AICD and summarized these signaling events in
the form of a network. This served as an innovative platform to
understand the abnormal CTL survival in T-LGL leukemia.
Multiple perturbations of signaling pathways in a disease can
result from deregulation of only a subset of these pathways.
However, identifying such a subset is difficult experimentally. In
this study, we assessed this question in T-LGL leukemia by using
a Boolean dynamic model. Through network simulations, we
revealed the hierarchy among deregulated nodes in terms of
determining other signaling abnormalities in leukemic T-LGL.
The simultaneously constitutive presence of IL-15 and PDGF
was shown to be sufficient to induce all know deregulations after
initial T cell activation.
Leukemic T-LGL are suggested to be antigen-primed (12),
long-term competent CTL (22), similar to terminally differen-
tiated effector memory cells (TEMRA) (23). It is worth noting that
IL-15 has been shown to be important for CTL activation and
generation of long-lived CD8 memory cells (24, 25). Both the
CD8 cells in the IL15 transgenic mice (26) and the IL15
transduced primary human CD8 cells (27) indeed showed
similar phenotypes as leukemic T-LGL. However, IL-15 alone
cannot fully inhibit the onset of apoptosis in our model (Fig. 2C).
Increased PDGF production occurs under inflammatory condi-
tions (28), and it has been shown to exert an inhibitory effect
toward CTL activation (29). We had found that LGL leukemia
is characterized by production of proinflammatory cytokines
(14). Here, we showed an increased level of PDGF in T-LGL
patient sera. Our model and the following validation revealed
that both pro- and anti-T cell activation signals are needed
simultaneously to maintain the competency and survival of CTL
in T-LGL leukemia. Our finding also suggests that provision of
IL-15 and PDGF may be a strategy to generate long-lived CTL
necessary for the development of virus and cancer vaccines.
Focusing on the effect of nodes on apoptosis in leukemic
T-LGL, we revealed nodes that determine the uncoupling of
CTL activation and AICD. We experimentally validated two
predicted key mediators: SPHK1 and NFB, the inhibition of
which induces apoptosis in leukemic T-LGL. It is worth noting
that although NFB has been suggested to inhibit apoptosis in
CTL (30), its function in maintaining long-term CTL survival
remains elusive. We validated the prediction that NFB is
downstream of PI3K and prevents the onset of apoptosis in
leukemic T-LGL through maintaining the expression of Mcl-1
independent of STAT3, another regulator of Mcl-1 in T-LGL
leukemia (3).
The confirmation of the constitutive NFB activation led to
the validation of the constitutive T-bet activation in T-LGL
leukemia predicted by our model. T-bet plays an important role
in coupling the effector and memory CD8 T cell fate (31). It
also inhibits the production of IL-2, which is known to be absent
in leukemic T-LGL and in TEMRA (23, 32). T-bet has been
related to multiple autoimmune diseases in humans (33–36).
T-LGL leukemia has important overlaps with autoimmune
disorders, particularly rheumatoid arthritis (2). The overexpres-
sion and constitutive activity of T-bet in leukemic T-LGL may
help to reveal the common pathogenesis between T-LGL leu-
kemia and other autoimmune diseases.
With the exponential increase of signaling pathway information,
it is becoming more difficult to determine pathway interactions in
a particular experimental system. In this study, we used network
analysis and Boolean modeling to investigate the signaling abnor-
malities in T-LGL leukemia. This systems biology approach was
able to maximize the use of the available pathway information and
to identify the key mediators of CTL survival, highlighting their
importance as potential therapeutic targets for T-LGL leukemia
0
0.2
0.4
0.6
0.8
1
0
5
10
15
20
25
Rounds of updating
Frequency of node
activation
0
0.01
0.02
0.03
0.04
0.05
Normal
T-LGL
Relative expression
*
3 4 6 9 10 11 12 13
Normal T-LGL
T-bet
GAPDH
T-bet
binding
Free
probe
T-LGL Normal
5 6 7 8 9
6 14 15 16 17
A
B
C
D
◊
IL-2
T-Bet
Fig. 6.
T-bet is overexpressed and constitutively
active in T-LGL leukemia PBMCs. (A) T-bet inhibits IL-2
expression when a T-LGL-like state is achieved. Based
on the Boolean logical rule ‘‘IL2* (NFKB or STAT3 or
NFAT) and not (TBET or Apoptosis)’’ (Table S4), T-bet
is the only negative regulator of IL-2 expression when
cells are still alive. Inhibiting T-bet (white squares)
after 15 rounds of updating results in IL-2 (white
diamonds) expression after achieving a T-LGL-like
state (200 simulations). (B) T-LGL leukemia PBMCs
(white squares, n 10) express 3.3-fold higher T-bet
mRNA compared with normal (black circles, n 5, *,
P 0.02) as assessed by real-time PCR. (C) T-bet pro-
tein expression in T-LGL leukemia and normal PBMCs.
Western blot assay result is representative of samples
from eight healthy donors and six T-LGL leukemia
patients. White space has been inserted to indicate
realigned gel lanes. (D) T-bet is constitutively active in
T-LGL leukemia patients. Nuclear extract from PBMCs
of five T-LGL leukemia patients and five healthy do-
nors were tested for their T-bet activity by using EMSA. T-bet exhibited high activity in most T-LGL leukemia patients but not in normal.
16312
www.pnas.orgcgidoi10.1073pnas.0806447105
Zhang et al.
and offering insights into CTL manipulation. This study confirms
the possibility of integrating normal and disease pathway informa-
tion into a single model that is powerful enough to reproduce a
clinically relevant complex process, useful for therapeutic purpose,
yet is constructed with only qualitative information.
Materials and Methods
Patient Consent. All patients met the clinical criteria of T-LGL (CD3) leukemia
with increased LGL counts and clonal T cell antigen receptor gene rearrange-
ment. None of the patients received treatment for LGL leukemia. Informed
consents were signed by all patients and age- and sex-matched healthy
individuals allowed the use of their cells for these experiments. Buffy coats
were obtained from Hershey Medical Center Blood Bank according to proto-
cols observed by Milton S. Hershey Medical Center, Hershey, PA.
Chemicals and Reagents. All chemical reagents and LY 294002 were purchased
from Sigma–Aldrich. AG 1296 and BAY 11–7082 was purchased from EMD
Bioscience. LightShift Chemiluminescent EMSA Kit was purchased from Pierce
Biotechnology. Human PDGF-BB Quantikine ELISA Kit was purchased from
R&D systems. Annexin V-PE Apoptosis Detection Kit-I and TransFactor Extrac-
tion Kit were purchased from BD Biosciences. Biotinylated and nonbiotin-
ylated DNA oligonucleotides were purchased from Integrated DNA Technol-
ogy. Anti-Mcl-1 and anti-T-bet antibodies were purchased from Santa Cruz
Biotechnology. Anti-AKT and anti-phospho-AKT (Ser-473) antibodies were
purchased from Cell Signaling Technology. Anti-GAPDH antibody was pur-
chased from Chemicon and Millipore. SPHK1 inhibitor I and II (SKI-I and SKI-II)
were kindly provided by Dr. Jong K. Yun.
ELISA. Sera from 22 T-LGL patients and 39 age- and sex-matched healthy
donors were prepared and analyzed according to manufacturer’s instruction,
as described (14).
Cell Culture and Apoptosis Assay. PBMCs were processed from blood samples
of patients and Buffy coats of normal donors, and apoptosis assays were
performed by using Annexin-V conjugated with phytoerythrocin (PE) and
7-amino-actinomycin-D staining as described (23). Each treatment was per-
formed and measured three times for one blood sample. Apoptosis was
calculated by using the following formula:
Percentage of specific apoptosis [(Annexin-V-PE positive cells in treat-
ment Annexin-V-PE positive cells in control) 100]/(100 Annexin-V-PE
positive cells in control)
Western Blot Assay. Cell lysates were prepared, protein concentration was
determined, and Western blot assay was performed as described (3).
EMSA. Nuclear and cytoplasmic extract from patient and normal PBMCs were
prepared by using TransFactor Extraction Kit according to manufacturer’s
instructions. Probes for NFB (5-GATCCGGCAGGGGAATCTCCCTCTC-3) (20),
STAT3 (5-CTTCATTTCCCGTAAATCCCTA) (3) and T-bet (5-AAAACTTGT-
GAAAATACGTAATCCTCAG-3) (34)were biotinylated at 5. Corresponding
nonbiotinylated oligonucleotides were used as competition oligonucleotides.
EMSA was performed by using the LightShift Chemiluminescent EMSA Kit
according to manufacturer’s instructions.
Real-Time PCR. Total RNA was processed as described (8). Primers specific for
T-bet (forward: 5-TGTGGTCCAAGTTTAATCAGCA-3; reverse: 5-TGACAG-
GAATGGGAACATCC-3) and glyceraldehyde-3-phosphate dehydro-genase
(GAPDH, forward: 5-GAGTCAACGGATTTGGTCGT-3; reverse: 5-TTGATTTTG-
GAGGGATCTCG-3) were used for real-time PCR and expression was quanti-
fied as described (8).
Computational Methods. Network simplification was performed with NET-
SYNTHESIS, a signal transduction network inference and simplification tool
(37, 38). The dynamic model was implemented in custom python code.
Additional details regarding computational methods are given in the SI
Text.
ACKNOWLEDGMENTS. We thank Nate Sheaffer and David Stanford for help
with acquisition and analysis of flow cytometry data and Lynn Ruiz, Kendall
Thomas, and Nancy Ruth Jarbadan for help with acquiring patient samples
and processing them. This work is supported by National Institutes of Health
Grant R01 CA 94872. Boolean model development in R.A’s group is supported
by National Science Foundation Grant CCF-0643529.
1. Klebanoff CA, Gattinoni L, Restifo NP (2006) CD8 T-cell memory in tumor immunol-
ogy and immunotherapy. Immunol Rev 211:214–224.
2. Sokol L, Loughran TP, Jr (2006) Large granular lymphocyte leukemia. Oncologist
11:263–273.
3. Epling-Burnette PK, et al. (2001) Inhibition of STAT3 signaling leads to apoptosis of
leukemic large granular lymphocytes and decreased Mcl-1 expression. J Clin Invest
107:351–362.
4. Schade AE, Powers JJ, Wlodarski MW, Maciejewski JP (2006) Phosphatidylinositol-3-
phosphate kinase pathway activation protects leukemic large granular lymphocytes
from undergoing homeostatic apoptosis. Blood 107:4834–4840.
5. Epling-Burnette PK, et al. (2004) ERK couples chronic survival of NK cells to constitu-
tively activated Ras in lymphoproliferative disease of granular lymphocytes (LDGL).
Oncogene 23:9220–9229.
6. Lamy T, Liu JH, Landowski TH, Dalton WS, Loughran TP, Jr (1998) Dysregulation of
CD95/CD95 ligand-apoptotic pathway in CD3 large granular lymphocyte leukemia.
Blood 92:4771–4777.
7. Krueger A, Fas SC, Baumann S, Krammer PH (2003) The role of CD95 in the regulation
of peripheral T-cell apoptosis. Immunol Rev 193:58–69.
8. Shah MV, et al. (2008) Molecular profiling of LGL leukemia reveals role of sphingolipid
signaling in survival of cytotoxic lymphocytes. Blood 112:770–781.
9. Christensen C, Thakar J, Albert R (2007) Systems-level insights into cellular regula-
tion: Inferring, analysing, and modelling intracellular networks. IET Syst Biol
1:61–77.
10. Li S, Assmann SM, Albert R (2006) Predicting essential components of signal transduc-
tion networks: A dynamic model of guard cell abscisic acid signaling. PLoS Biol 4:e312.
11. Chaves M, Albert R, Sontag ED (2005) Robustness and fragility of Boolean models for
genetic regulatory networks. J Theor Biol 235(3):431–449.
12. Wlodarski MW, et al. (2005) Pathologic clonal cytotoxic T-cell responses: Nonrandom
nature of the T-cell-receptor restriction in large granular lymphocyte leukemia. Blood
106:2769–2780.
13. Zambello R, et al. (1997) Interleukin-15 triggers the proliferation and cytotoxicity of
granular lymphocytes in patients with lymphoproliferative disease of granular lym-
phocytes. Blood 89:201–211.
14. Kothapalli R, et al. (2005) Constitutive production of proinflammatory cytokines
RANTES, MIP-1beta and IL-18 characterizes LGL leukemia. Int J Oncol 26:529–535.
15. Heldin C-H, Westermark B (1999) Mechanism of action and in vivo role of platelet-
derived growth factor. Physiol Rev 79:1283–1316.
16. French KJ, et al. (2003) Discovery and evaluation of inhibitors of human sphingosine
kinase. Cancer Res 63:5962–5969.
17. French KJ, et al. (2006) Antitumor activity of sphingosine kinase inhibitors. J Pharmacol
Exp Ther 318:596–603.
18. Hoffmann A, Baltimore D (2006) Circuitry of nuclear factor B signaling. Immunol Rev
210:171–186.
19. Pierce JW, et al. (1997) Novel Inhibitors of Cytokine-induced I B Phosphorylation and
Endothelial Cell Adhesion Molecule Expression Show Anti-inflammatory Effects in
Vivo. J Biol Chem 272:21096–21103.
20. Mori N, et al. (2002) Bay 11–7082 inhibits transcription factor NF-kappa B and induces
apoptosis of HTLV-I-infected T-cell lines and primary adult T-cell leukemia cells. Blood
100:1828–1834.
21. Loughran TP, Jr., Aprile JA, Ruscetti FW (1990) Anti-CD3 monoclonal antibody-
mediated cytotoxicity occurs through an interleukin-2-independent pathway in CD3
large granular lymphocytes. Blood 75:935–940.
22. Kothapalli R, et al. (2003) Constitutive expression of cytotoxic proteases and down-
regulation of protease inhibitors in LGL leukemia. Int J Oncol 22:33–39.
23. Yang J, et al. (2008) Antigen activation and impaired Fas-induced death-inducing
signaling complex formation in T-large-granular lymphocyte leukemia. Blood
111:1610–1616.
24. Liu K, Catalfamo M, Li Y, Henkart PA, Weng N-p (2002) IL-15 mimics T cell receptor
crosslinking in the induction of cellular proliferation, gene expression, and cytotoxicity
in CD8 memory T cells. Proc Natl Acad Sci USA 99:6192–6197.
25. Weng N-P, Liu K, Catalfamo M, Li YU, Henkart PA (2002) IL-15 is a growth factor and
an activator of CD8 memory T cells. Ann NY Acad Sci 975:46–56.
26. Fehniger TA, et al. (2001) Fatal leukemia in interleukin 15 transgenic mice follows early
expansions in natural killer and memory phenotype CD8 T cells. J Exp Med 193:219–
232.
27. Hsu C, et al. (2007) Cytokine-independent growth and clonal expansion of a primary
human CD8 T-cell clone following retroviral transduction with the IL-15 gene. Blood
109:5168–5177.
28. Mannaioni PF, Di Bello MG, Masini E (1997) Platelets and inflammation: Role of
platelet-derived growth factor, adhesion molecules and histamine. Inflammation Res
46:4–18.
29. Daynes RA, Dowell T, Araneo BA (1991) Platelet-derived growth factor is a potent
biologic response modifier of T cells. J Exp Med 174:1323–1333.
30. Hayden MS, West AP, Ghosh S (2006) NF-B and the immune response. Oncogene
25:6758–6780.
31. Intlekofer AM, et al. (2005) Effector and memory CD8 T cell fate coupled by T-bet and
eomesodermin. Nat Immunol 6:1236–1244.
32. Hamann D, et al. (1997) Phenotypic and functional separation of memory and effector
human CD8 T cells. J Exp Med 186:1407–1418.
33. Nath N, Prasad R, Giri S, Singh AK, Singh I (2006) T-bet is essential for the progression
of experimental autoimmune encephalomyelitis. Immunology 118:384–391.
34. Solomou EE, Keyvanfar K, Young NS (2006) T-bet, a Th1 transcription factor, is
up-regulated in T cells from patients with aplastic anemia. Blood 107:3983–3991.
35. Juedes AE, Rodrigo E, Togher L, Glimcher LH, von Herrath MG (2004) T-bet controls
autoaggressive CD8 lymphocyte responses in type 1 diabetes. J Exp Med 199:1153–1162.
36. Lovett-Racke AE, et al. (2004) Silencing T-bet defines a critical role in the differentia-
tion of autoreactive T lymphocytes. Immunity 21:719–731.
37. Albert R, et al. (2007) A novel method for signal transduction network inference from
indirect experimental evidence. J Comput Bio 14:927–949.
38. Kachalo S, Zhang R, Sontag E, Albert R, DasGupta B (2008) NET-SYNTHESIS: A software
for synthesis, inference and simplification of signal transduction networks. Bioinfor-
matics 24:293–295.
Zhang et al.
PNAS
October 21, 2008
vol. 105
no. 42
16313
MEDICAL SCIENCES
|
18852469
|
FasT = ( ( NFKB ) AND NOT ( Apoptosis ) )
sFas = ( ( FasT ) AND NOT ( Apoptosis ) )
IL2 = ( ( ( STAT3 ) AND NOT ( Apoptosis ) ) AND NOT ( TBET ) ) OR ( ( ( NFAT ) AND NOT ( Apoptosis ) ) AND NOT ( TBET ) ) OR ( ( ( NFKB ) AND NOT ( Apoptosis ) ) AND NOT ( TBET ) )
NFKB = ( ( PI3K ) AND NOT ( Apoptosis ) ) OR ( ( TPL2 ) AND NOT ( Apoptosis ) ) OR ( ( FLIP AND ( ( ( IAP AND TRADD ) ) ) ) AND NOT ( Apoptosis ) )
TPL2 = ( ( PI3K AND ( ( ( TNF ) ) ) ) AND NOT ( Apoptosis ) ) OR ( ( TAX ) AND NOT ( Apoptosis ) )
A20 = ( ( NFKB ) AND NOT ( Apoptosis ) )
GPCR = ( ( S1P ) AND NOT ( Apoptosis ) )
IFNGT = ( ( TBET ) AND NOT ( Apoptosis ) ) OR ( ( STAT3 ) AND NOT ( Apoptosis ) ) OR ( ( NFAT ) AND NOT ( Apoptosis ) )
GZMB = ( ( TBET ) AND NOT ( Apoptosis ) ) OR ( ( CREB AND ( ( ( IFNG ) ) ) ) AND NOT ( Apoptosis ) )
TCR = ( ( ( Stimuli ) AND NOT ( CTLA4 ) ) AND NOT ( Apoptosis ) )
IL2RBT = ( ( ERK AND ( ( ( TBET ) ) ) ) AND NOT ( Apoptosis ) )
TRADD = ( ( ( ( TNF ) AND NOT ( IAP ) ) AND NOT ( Apoptosis ) ) AND NOT ( A20 ) )
PI3K = ( ( PDGFR ) AND NOT ( Apoptosis ) ) OR ( ( RAS ) AND NOT ( Apoptosis ) )
FYN = ( ( TCR ) AND NOT ( Apoptosis ) ) OR ( ( IL2RB ) AND NOT ( Apoptosis ) )
IL2RA = ( ( ( IL2 AND ( ( ( IL2RAT ) ) ) ) AND NOT ( IL2RA ) ) AND NOT ( Apoptosis ) )
GRB2 = ( ( IL2RB ) AND NOT ( Apoptosis ) ) OR ( ( ZAP70 ) AND NOT ( Apoptosis ) )
BID = ( ( ( ( Caspase ) AND NOT ( BclxL ) ) AND NOT ( MCL1 ) ) AND NOT ( Apoptosis ) ) OR ( ( ( ( GZMB ) AND NOT ( BclxL ) ) AND NOT ( MCL1 ) ) AND NOT ( Apoptosis ) )
Caspase = ( ( DISC ) AND NOT ( Apoptosis ) ) OR ( ( ( GZMB AND ( ( ( BID ) ) ) ) AND NOT ( Apoptosis ) ) AND NOT ( IAP ) ) OR ( ( ( TRADD AND ( ( ( BID ) ) ) ) AND NOT ( Apoptosis ) ) AND NOT ( IAP ) )
ZAP70 = ( ( ( LCK ) AND NOT ( FYN ) ) AND NOT ( Apoptosis ) )
CTLA4 = ( ( TCR ) AND NOT ( Apoptosis ) )
LCK = ( ( CD45 ) AND NOT ( Apoptosis ) ) OR ( ( ( TCR ) AND NOT ( Apoptosis ) ) AND NOT ( ZAP70 ) ) OR ( ( ( IL2RB ) AND NOT ( Apoptosis ) ) AND NOT ( ZAP70 ) )
SOCS = ( ( ( ( JAK ) AND NOT ( Apoptosis ) ) AND NOT ( IL2 ) ) AND NOT ( IL15 ) )
STAT3 = ( ( JAK ) AND NOT ( Apoptosis ) )
Apoptosis = ( Caspase ) OR ( Apoptosis )
IL2RB = ( ( IL2RBT AND ( ( ( IL2 OR IL15 ) ) ) ) AND NOT ( Apoptosis ) )
Fas = ( ( ( FasT AND ( ( ( FasL ) ) ) ) AND NOT ( sFas ) ) AND NOT ( Apoptosis ) )
Cytoskeleton_signaling = ( ( FYN ) AND NOT ( Apoptosis ) )
FLIP = ( ( ( NFKB ) AND NOT ( DISC ) ) AND NOT ( Apoptosis ) ) OR ( ( ( CREB AND ( ( ( IFNG ) ) ) ) AND NOT ( DISC ) ) AND NOT ( Apoptosis ) )
JAK = ( ( ( ( IFNG ) AND NOT ( Apoptosis ) ) AND NOT ( CD45 ) ) AND NOT ( SOCS ) ) OR ( ( ( ( RANTES ) AND NOT ( Apoptosis ) ) AND NOT ( CD45 ) ) AND NOT ( SOCS ) ) OR ( ( ( ( IL2RA ) AND NOT ( Apoptosis ) ) AND NOT ( CD45 ) ) AND NOT ( SOCS ) ) OR ( ( ( ( IL2RB ) AND NOT ( Apoptosis ) ) AND NOT ( CD45 ) ) AND NOT ( SOCS ) )
IAP = ( ( ( NFKB ) AND NOT ( Apoptosis ) ) AND NOT ( BID ) )
FasL = ( ( ERK ) AND NOT ( Apoptosis ) ) OR ( ( NFAT ) AND NOT ( Apoptosis ) ) OR ( ( NFKB ) AND NOT ( Apoptosis ) ) OR ( ( STAT3 ) AND NOT ( Apoptosis ) )
S1P = ( ( ( SPHK1 ) AND NOT ( Apoptosis ) ) AND NOT ( Ceramide ) )
SPHK1 = ( ( PDGFR ) AND NOT ( Apoptosis ) )
ERK = ( ( MEK AND ( ( ( PI3K ) ) ) ) AND NOT ( Apoptosis ) )
Ceramide = ( ( ( Fas ) AND NOT ( S1P ) ) AND NOT ( Apoptosis ) )
MCL1 = ( ( IL2RB AND ( ( ( PI3K AND NFKB AND STAT3 ) ) ) ) AND NOT ( Apoptosis ) )
P2 = ( ( ( IFNG ) AND NOT ( Stimuli2 ) ) AND NOT ( Apoptosis ) ) OR ( ( ( P2 ) AND NOT ( Stimuli2 ) ) AND NOT ( Apoptosis ) )
RANTES = ( ( NFKB ) AND NOT ( Apoptosis ) )
NFAT = ( ( PI3K ) AND NOT ( Apoptosis ) )
GAP = ( ( ( ( PDGFR AND ( ( ( GAP ) ) ) ) AND NOT ( Apoptosis ) ) AND NOT ( IL2 ) ) AND NOT ( IL15 ) ) OR ( ( ( ( RAS ) AND NOT ( Apoptosis ) ) AND NOT ( IL2 ) ) AND NOT ( IL15 ) )
SMAD = ( ( GPCR ) AND NOT ( Apoptosis ) )
IFNG = ( ( ( ( IL15 AND ( ( ( IFNGT ) ) ) ) AND NOT ( P2 ) ) AND NOT ( Apoptosis ) ) AND NOT ( SMAD ) ) OR ( ( ( ( IL2 AND ( ( ( IFNGT ) ) ) ) AND NOT ( P2 ) ) AND NOT ( Apoptosis ) ) AND NOT ( SMAD ) ) OR ( ( ( ( Stimuli AND ( ( ( IFNGT ) ) ) ) AND NOT ( P2 ) ) AND NOT ( Apoptosis ) ) AND NOT ( SMAD ) )
TNF = ( ( NFKB ) AND NOT ( Apoptosis ) )
CREB = ( ( ERK AND ( ( ( IFN ) ) ) ) AND NOT ( Apoptosis ) )
P27 = ( ( STAT3 ) AND NOT ( Apoptosis ) )
Proliferation = ( ( ( STAT3 ) AND NOT ( P27 ) ) AND NOT ( Apoptosis ) )
TBET = ( ( TBET ) AND NOT ( Apoptosis ) ) OR ( ( JAK ) AND NOT ( Apoptosis ) )
MEK = ( ( RAS ) AND NOT ( Apoptosis ) )
RAS = ( ( ( GRB2 ) AND NOT ( Apoptosis ) ) AND NOT ( GAP ) ) OR ( ( ( PLCG1 ) AND NOT ( Apoptosis ) ) AND NOT ( GAP ) )
PLCG1 = ( ( GRB2 ) AND NOT ( Apoptosis ) ) OR ( ( PDGFR ) AND NOT ( Apoptosis ) )
PDGFR = ( ( PDGF ) AND NOT ( Apoptosis ) ) OR ( ( S1P ) AND NOT ( Apoptosis ) )
BclxL = ( ( ( ( ( STAT3 ) AND NOT ( BID ) ) AND NOT ( Apoptosis ) ) AND NOT ( GZMB ) ) AND NOT ( DISC ) ) OR ( ( ( ( ( NFKB ) AND NOT ( BID ) ) AND NOT ( Apoptosis ) ) AND NOT ( GZMB ) ) AND NOT ( DISC ) )
IL2RAT = ( ( IL2 AND ( ( ( NFKB OR STAT3 ) ) ) ) AND NOT ( Apoptosis ) )
DISC = ( ( FasT AND ( ( ( Fas ) AND ( ( ( NOT FLIP ) ) ) ) OR ( ( Fas AND IL2 ) ) OR ( ( Ceramide ) ) ) ) AND NOT ( Apoptosis ) )
|
BioMed Central
Page 1 of 14
(page number not for citation purposes)
BMC Systems Biology
Open Access
Research article
Dynamical modeling of the cholesterol regulatory pathway with
Boolean networks
Gwenael Kervizic* and Laurent Corcos
Address: Inserm U613, Faculté de Médecine, Université de Bretagne Occidentale, 29238 Brest cedex, FRANCE
Email: Gwenael Kervizic* - gwenael.kervizic@univ-brest.fr; Laurent Corcos - laurent.corcos@univ-brest.fr
* Corresponding author
Abstract
Background: Qualitative dynamics of small gene regulatory networks have been studied in quite
some details both with synchronous and asynchronous analysis. However, both methods have their
drawbacks: synchronous analysis leads to spurious attractors and asynchronous analysis lacks
computational efficiency, which is a problem to simulate large networks. We addressed this
question through the analysis of a major biosynthesis pathway. Indeed the cholesterol synthesis
pathway plays a pivotal role in dislypidemia and, ultimately, in cancer through intermediates such
as mevalonate, farnesyl pyrophosphate and geranyl geranyl pyrophosphate, but no dynamic model
of this pathway has been proposed until now.
Results: We set up a computational framework to dynamically analyze large biological networks.
This framework associates a classical and computationally efficient synchronous Boolean analysis
with a newly introduced method based on Markov chains, which identifies spurious cycles among
the results of the synchronous simulation. Based on this method, we present here the results of
the analysis of the cholesterol biosynthesis pathway and its physiological regulation by the Sterol
Response Element Binding Proteins (SREBPs), as well as the modeling of the action of statins,
inhibitor drugs, on this pathway. The in silico experiments show the blockade of the cholesterol
endogenous synthesis by statins and its regulation by SREPBs, in full agreement with the known
biochemical features of the pathway.
Conclusion: We believe that the method described here to identify spurious cycles opens new
routes to compute large and biologically relevant models, thanks to the computational efficiency of
synchronous simulation.
Furthermore, to the best of our knowledge, we present here the first dynamic systems biology
model of the human cholesterol pathway and several of its key regulatory control elements, hoping
it would provide a good basis to perform in silico experiments and confront the resulting properties
with published and experimental data. The model of the cholesterol pathway and its regulation,
along with Boolean formulae used for simulation are available on our web site http://
Bioinformaticsu613.free.fr. Graphical results of the simulation are also shown online. The SBML
model is available in the BioModels database http://www.ebi.ac.uk/biomodels/ with submission ID:
MODEL0568648427.
Published: 24 November 2008
BMC Systems Biology 2008, 2:99
doi:10.1186/1752-0509-2-99
Received: 11 April 2008
Accepted: 24 November 2008
This article is available from: http://www.biomedcentral.com/1752-0509/2/99
© 2008 Kervizic and Corcos; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 2 of 14
(page number not for citation purposes)
Background
Systems biology
Systems biology is an emerging scientific field that inte-
grates large sets of biological data derived from experi-
mental and computational approaches. In this new
paradigm, we no longer study entities of biological sys-
tems separately, but as a whole. Hence, large data sets can
be translated into sets of links representative of the inter-
actions of species from within single or multiple path-
ways. In fact, elementary functions in those systems are
the result of the inherent characteristics of the specific ele-
ments involved and the interactions they are engaged in
within the systems [1]. In biological or biomedical mat-
ters, modeling activities are strongly linked to the nature
and amount of available data on the model. Furthermore,
computational studies in systems biology rely on different
formalisms that are intimately connected to the level of
knowledge one has of a biological system. In the present
study, the cholesterol synthesis pathway, including most
of its associated reactions, is analyzed to address the effect
of either activators or inhibitors. Hence, blockade can be
attained by targeting the HMG-CoA reductase, the rate-
limiting enzyme of the mevalonate pathway, with statins,
widely used hypocholesterolemic drugs. Alternatively,
activation of the pathway can be triggered by Sterol
Response Element Binding Proteins (SREBPs), as part of a
compensatory feedback mechanism. Moreover, to better
analyze this pathway including both enzymatic reactions
and gene regulatory networks, we will focus on the
Boolean networks formalism, particularly suitable to
delineate dynamic properties from qualitative informa-
tion on regulatory interactions [2,3].
Boolean formalism for qualitative modeling and
simulation
A model or simulation of a biological network is said to
be qualitative when each entity of this model is repre-
sented by a variable having a finite set of possible values.
We can note here that the possible values that can be
taken by the variable are not necessarily linearly correlated
to the concentration of the represented species. Those val-
ues represent qualitative states of the entities from the net-
work. In the formalism of Boolean networks, the state of
a species is described by a Boolean variable, which value
is either 1 if the species is active (i.e. its activity is detecta-
ble, in biological terms) or 0 if inactive (its activity is
undetectable). Moreover, a Boolean function allows to
compute the state of a species at time t + 1, knowing the
states of k other species at time t. If we denote by xi the
state of species i and by bi(x(t)) the associated Boolean
function, we get the following equations for the dynamics
of the Boolean network:
xi(t + 1) = bi(x(t)), 1 ≤ i ≤ n
(1)
We can note here that the Boolean formalism allows us to
model various biological systems such as gene regulatory
networks and metabolic networks whose entities have
very different timescales.
Construction of a Boolean network: modeling inhibition
and activation
Let us detail how inhibitions and activations should be
modeled in the Boolean network formalism.
• Inhibition: if A is an enzyme that produces a compound
B but can be inhibited by compound C, then the Boolean
function that predicts the presence of B at time t + 1 will
be: B(t + 1) = A(t) AND NOT(C(t))
• Activation: if A is a precursor of B and the reaction of
transformation of A to B is catalyzed by enzyme C, then
the Boolean function that predicts the presence of B at
time t + 1 will be: B(t + 1) = A(t) AND C(t)
Here is a simple example with 4 genes (A, B, C, D) and the
4 following Boolean functions:
• A(t + 1) = NOT (D(t))
• B(t + 1) = NOT (A(t))
• C(t + 1) = A(t) OR B(t)
• D(t + 1) = NOT (C(t))
The graphical representation of this network can be seen
in figure 1.
Example of a simple regulatory network
Figure 1
Example of a simple regulatory network. Graphical
representation of a regulatory network with 4 genes (A, B,
C, D). Its full dynamics is described in the associated Boolean
functions.
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 3 of 14
(page number not for citation purposes)
Synchronous and asynchronous paradigms in the Boolean
formalism
In Boolean simulations, there are two main paradigms
where conception of time and transition between states
differs.
• The simplest one is the synchronous simulation. At each
step of clock (time is moving discretely in Boolean simu-
lations) all the decrease or increase calls are realized
simultaneously. This approach is computationally effi-
cient, but might lead to simulation artifacts such as spuri-
ous cycles [4,5], which are cycles that do not appear in
asynchronous simulation.
• In asynchronous simulation, only one transition occurs
at each clock step. Thus, the same reaction can occur sev-
eral times before another one is completed, which enables
the simulation of biological systems that contain slow
and fast kinetics (equivalent to a stiff system in the Ordi-
nary Differential Equations paradigm).
It is worthwhile to note that the steady states, which cor-
respond to some phenotypes, are the same in those two
paradigms. However, some dynamic behaviors can be
very different.
To sum up, synchronous simulations have fewer mode-
ling power but are more computationally efficient while
asynchronous simulations are able to predict a wider
range of biological behaviors but their exhaustive compu-
tation becomes intractable for large biological systems
[6,7].
In the synchronous paradigm simulation, our simple reg-
ulatory network gives the results partially shown in table
1. The study of this truth table shows that {1010} is a
steady state (or point attractor, or equilibrium) and that
{0010, 1100, 1011} is a state cycle (or dynamic attractor,
or cyclic attractor). This becomes more evident when con-
verting this network into a finite state machine as shown
in figure 2. The state colored in green corresponds to the
steady state and the states colored in red correspond to the
state cycle.
In the asynchronous paradigm simulation, our simple
regulatory network gives the finite state machine shown in
figure 3. The state colored in green corresponds to the
Table 1: Fragment of the truth table obtained from our simple
regulatory network.
(ABCD)
t
0000
0001
0010
0011
0100
0101
...
t + 1
1101
0101
1100
0100
1111
0111
...
For each initial array of values (initial state) at time t, the new array of
values, obtained by evaluating the system through the Boolean
functions, is shown on the second line (t + 1).
Finite state machine of our regulatory network taken as an
example in synchronous simulation
Figure 2
Finite state machine of our regulatory network taken
as an example in synchronous simulation. The state
[1010] colored in green corresponds to a steady state. It has
5 states and itself in its basin of attraction (i.e. the states
whose trajectory during the simulation lead to this steady
state). The 3 states [0010], [1100], [1011] colored in red
correspond to a state cycle. They have 7 states and them-
selves in their basin of attraction. All the state space is shown
in the figure.
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 4 of 14
(page number not for citation purposes)
steady state in both synchronous and asynchronous sim-
ulations. We recall that steady states are obviously always
the same in synchronous and asynchronous simulations.
The states colored in red are the states which correspond
to the state cycle in synchronous simulation. The purple
arrows propose one way -among many possible- to reach
cyclically those states. Note that, for some regulatory net-
works, it is not possible in asynchronous simulation to
reach cyclically states which form a state cycle in synchro-
nous simulation. This becomes obvious when looking at
the synchronous and asynchronous simulation results of
a simple negative feedback loop of size 3. The synchro-
nous state cycle {010, 101} does not exist in asynchro-
nous simulation. These results are shown on our web site
http://bioinformaticsu613.free.fr/simpleloopsn3.html.
Furthermore, we can have an intuition that this purple tra-
jectory is in some way unstable because while cycling
through the 3 states {0010, 1100, 1011}, the system
could have gone through many transitions that lead to the
steady state 1010.
The cholesterol biosynthesis pathway
Cholesterol is an important constituent of mammalian
cell membranes. It maintains their fluidity and allows
other molecules playing important biological roles, like
glycoproteins, to anchor to the membrane compartment.
It is also the precursor of fat-soluble vitamins, including
vitamins A, D, E and K and of various steroids hormones,
such as cortisol, aldosterone, progesterone, the various
estrogens and testosterone. It comes for about one third
from the dietary intake and for about two thirds from
endogenous synthesis from unburned food metabolites.
Its synthesis starts from acetyl CoA, through what is often
called the HMG-CoA reductase pathway. It occurs in
many cells and tissues, but with higher rates in the intes-
tines, adrenal glands, reproductive organs and liver. Cho-
lesterol synthesis is orchestrated by a protein complex
formed by the Sterol Regulatory Element Binding Protein
(SREBP), the SREBP-cleavage activating protein (SCAP)
and the insulin-induced gene 1 (Insig) [8-10]. This com-
plex is maintained in a repressed state located in the endo-
plasmic reticulum (ER). When the cholesterol level is low,
Insig1 interaction with SREBP-SCAP complex is relieved
allowing SREBP-SCAP to migrate to the Golgi apparatus
where SREBP is cleaved by two proteases called S1P and
S2P. Once SREBP is matured, it migrates to the nucleus
and acts as a transcription factor upon binding to sterol
regulatory elements (SRE) to activate the genes coding for
the main enzymes of the HMG-CoA reductase pathway
(e.g. HMG-CoA synthase, HMG-CoA reductase, FPP syn-
thase, CYP51). The synthesis of cholesterol can be regu-
lated by drugs such as HMG-CoA reductase inhibitors,
among which the most potent belong to the statins family
[11-14]. They lower cholesterol by inhibiting the enzyme
HMG-CoA reductase, which is rate-limiting.
Effects of statins on cancer activated pathways
Therefore statins are known lipopenic drugs, but they are
also drug candidates against cancer [15]. Intermediate
molecules in the HMG-CoA reductase pathway undergo
important biochemical reactions of prenylation whose
blocking will inactivate several intracellular transduction
pathways that involve Ras, Rho and small G proteins [16-
Finite state machine of our regulatory network taken as an
example in asynchronous simulation
Figure 3
Finite state machine of our regulatory network taken
as an example in asynchronous simulation. The state
[1010] colored in green also corresponds to a steady state in
the asynchronous simulation. (All the steady states are the
same in synchronous and asynchronous simulations) The 3
states [0010], [1100], [1011] which correspond to a state
cycle in the synchronous simulation are still colored in red,
but this figure clearly shows that they do not correspond to
a state cycle in the asynchronous simulation. Actually, even if
there are several paths enabling to reach cyclically those
three states (one of those paths is indicated with purple
arrows), there are also several paths leading to the steady
state from which there is no path back to those 3 states.
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 5 of 14
(page number not for citation purposes)
19]. Hence, statins can block Ras activation, which occurs
in 30% of human tumours. Experimentally, statins can
stop cell growth by blocking cells at the G1 or G2/M
stages, or induce apoptosis in several cancer cell types
[20]. Important results have also been obtained using
rodent models where neuroblastoma, colic cancer and
melanoma have regressed under the effect of lovastatin.
Moreover, the combination of classical antineoplastic
drugs, like DNA topoisomerase inhibitors, and statins
increases tumour cell killing [21,22].
In this paper, we first focus on the synchronous formalism
enabling us to compute our large model of cholesterol
regulatory pathway. We next propose a methodology
based on asynchronous formalism and of Markov chains
to overcome one of its limitations: the appearance of spu-
rious cycles.
Methods
Boolean modeling of the human cholesterol regulatory
pathway
The model shown in figure 4 has been made using data
from the literature [23-25]. It is composed of the choles-
terol synthesis pathway and its regulation by SREBPs.
After a few simplifications aimed at reducing the state
space size, our model includes 33 species (genes, mRNAs,
proteins, biochemical intermediates and statins). In par-
ticular, we assume the SREBP-SCAP-Insig1 complex to be
always present in the membranes of the endoplasmic
reticulum (ER). Therefore, in our model, the SREBP-SCAP
complex can be either present in the ER if the cholesterol
level is high, or absent if the cholesterol level is below the
physiologically relevant threshold, a situation that occurs
following its dissociation from Insig. We also assume that
S1p and S2p are always present in the membranes of the
Golgi apparatus. Hence, in our model, a non-fully
matured SREBP protein, called here pSREBP (for precur-
sor-SREBP) is automatically produced if SREBP-SCAP is
present. Likewise, matured SREBP, called mSREBP, is
automatically derived from pSREBP and then migrates to
the nucleus to enhance transcription of the genes from the
HMG-CoA reductase pathway when cholesterol levels are
perceived as insufficient.
Target genes will then be transcribed into their respective
mRNA, which will be translated into the corresponding
enzymes. The endogenous cholesterol synthesis starts
with acetyl-CoA which can, in our model, be either
present, or absent in case of deficiency. Acetyl-CoA com-
bines with itself to give CoA-SH and acetoacetyl-CoA.
Acetyl-CoA reacts then with acetoacetyl-CoA to give
HMG-CoA (3-Hydroxy-3-methylglutaryl CoA). This reac-
tion is catalyzed by the HMG-CoA synthase. Therefore,
the Boolean formula that describes the evolution of
HMG-CoA is:
HMG_CoA(t+1) = Acetoacetyl_CoA(t) AND
Acetyl_CoA(t) AND HMG_CoA_Synthase(t)
as expressed in the formalism of equation (1).
We assume that NADPH and H+ are always present and
we have chosen not to represent them in our model. Thus,
in the presence of HMG-CoA reductase, HMG-CoA will
produce mevalonic acid. ATP is also considered to be
present in sufficient amounts so that mevalonic acid will
transform into mevalonyl pyrophosphate, which will
then transform into isopentenyl pyrophosphate.
Isopentenyl pyrophosphate will give dimethyl allyl pyro-
phosphate, and then combine with its own product to
form geranyl pyrophosphate. This last one will combine
with isopentenyl pyrophosphate to give farnesyl pyro-
phosphate. Farnesyl pyrophosphate will lose two inor-
ganic phosphates and one H+ ion to give presqualene
pyrophosphate that will get two hydrogens from NADPH
and H+ and lose two more inorganic phosphates to trans-
form into squalene.
Since we assume NAPDH and H+ to be always present in
enough quantity, farnesyl pyrophosphate will automati-
cally give squalene and presqualene pyrophosphate
which, as an intermediate of the reaction, is not men-
tioned.
The ring closure of squalene produces lanosterol. We have
then omitted several transitions and jumped from the
lanosterol to desmosterol or 7-dehydrocholesterol, which
both give cholesterol. The Boolean formula that describes
the formation of cholesterol from either desmosterol or 7-
dehydrocholesterol is:
Cholesterol(t+1) = Desmosterol(t) OR
7_dehydrocholesterol(t)
Perturbations of the model such as blockade of the HMG-
CoA reductase by statins, widely used hypocholestero-
lemic drugs, can be readily modeled. In the case of statins
and HMG-CoA reductase, the Boolean formula is:
HMG_CoA_Reductase(t+1) =
HMG_CoA_Reductase_RNA(t) AND NOT(Statins(t))
Encoding the model
At each time of the simulation the state of the targeted
biological system is represented by a Boolean vector. Each
coordinate represents a species in the pathway.
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 6 of 14
(page number not for citation purposes)
Cholesterol Regulatory Pathway
Figure 4
Cholesterol Regulatory Pathway. The cholesterol biosynthesis pathway is shown on this figure starting by its precursor
Acetyl-CoA. Several of the key enzymes of this pathway regulated by SREBPs are also shown. The genetic regulation initiated
by the effect of cholesterol on the Insig-SREBP-SCAP complex can be seen on the top of this figure. The action of statins,
widely used hypolipidemic drugs, on the HMG-CoA-Reductase enzyme is also captured in this graph. Species of the model are
grouped into cellular compartments as nucleus, golgi apparatus and endoplasmic reticulum. Default cellular compartment is the
cytosol. This graphical representation has been prepared with CellDesigner [60,61].
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 7 of 14
(page number not for citation purposes)
The evolution function takes a Boolean vector represent-
ing the state of the model at time t and returns a Boolean
vector representing the state of the model at time t + 1.
Storing the model
The study of those pathways is greatly facilitated when the
models are stored in a computer-readable format allowing
representation of a biological system. Such formats have
already been proposed like the Systems Biology Markup
Language (SBML) [26,27]. SBML files are static represen-
tations of biological systems that contain species, reac-
tions, kinetic laws and possible annotations. SBML
implements the XML (Extensible Markup Language)
standard and is now internationally supported and widely
used. It allows models of biological systems to be stored
in public or private databases. However, it has been
designed for the purpose of ODE (Ordinary Differential
Equations) simulations and thus needs some adaptations
to be used for Boolean simulations, like the possibility to
store Boolean formulae. For our purposes, we have used
SBML files with additional <BooleanLaws> </BooleanLaws>
tags which are stored within reaction tags in an annotated
section to remain compatible with SBML standard. This
SBML can be downloaded from the BioModels database
http://www.ebi.ac.uk/biomodels/[28] and has the follow-
ing submission ID: MODEL0568648427.
Simulation: point and cyclic attractors
In a synchronous simulation, every trajectory converges to
an attractor. Indeed, as the state space is finite (its size is
2N with N the number of species in the model), if we keep
the simulation running long enough it will eventually
come back to an already reached state. At that point, the
trajectory becomes periodic because the simulation is
deterministic on a finite state space.
If the periodic part of the trajectory is of size one, we call
the state a point attractor. When the system loops infi-
nitely through several states, we call the set of these states
a cycle attractor.
While non-attractor states are transient and visited at most
once on any network trajectory, states within an attractor
cycle or point are reached infinitely often. Thus, attractors
are often identified with phenotypes [2,3]. Considering
that a phenotype is an observable state, therefore stable, of
an organism or a cell, real biological systems are typically
assumed to have short attractor cycles [29].
The state-space explosion problem
In order to fully analyze the model with a simple simula-
tion approach, we would need to simulate every state of
the state-space. But the size of this space grows exponen-
tially with the number of species and thus the computa-
tion of the trajectories starting from all possible states will
rapidly become too costly. Thus, we have decided, as a
first step prior to a formal analysis, to use a random gen-
erator in order to choose a subset of start states signifi-
cantly smaller than the whole state-space and uniformly
distributed in this space. We have also taken advantage of
multiple processors computing in order to cover the max-
imum of the state space with a minimum of time. Algo-
rithms developed for Boolean simulation are very well
suited for parallel execution. For example the set of start
states used for simulation can easily be divided into sub-
sets and simulation can be run independently from those
start subsets. However, we think that parallelization is not
sufficient to overcome the combinatorial explosion.
Indeed, in order to add a species to a model, the comput-
ing capabilities must be multiplied by two.
Relative importance of cycles: A Markovian approach
based on asynchronous perturbations
We propose here a methodology based on Markov proc-
esses that computes the stability of cycles. Markov proc-
esses have already been used for controlling and analyzing
gene networks [30,31]. Our approach differs from what is
done in Probabilistic Boolean Networks (PBN) by the
choice of the state space: instead of classically using the
state space of the system itself (i.e. the 2N possible values
of the state vector), we will use the set of state cycles and
equilibria which is typically much smaller [2,3]. This
allows us to compute Markov chain-based algorithms on
large biological systems and thus to take into account the
substantial and still growing amount of data we have on
those networks and pathways. However, as a price for scal-
ability and unlike PBN, the framework of synchronous
Boolean networks does not represent the possible stochas-
ticity of state transitions. Let S be the set of results of our
Boolean analysis (S is composed of point attractors and
cyclic attractors). S is the state-space of the finite discrete
time-homogeneous Markov chain we want to study. In
order to define the transitions of this Markov chain, we
apply a perturbation on each cycle C in S. C has k states (k
can be 1 in the case of a point attractor), say (s1, s2, ..., sk).
For each state si of the cycle (s1, s2, ..., sk) the perturbation
consists in reevaluating each species by its own Boolean
function triggered asynchronously. Thus we obtain N new
states (si,1, si,2, ..., si,N) for each state in C. When we perturb
every state with every perturbation, we obtain a new set of
perturbed states of size kN.
We can note here that some of the states in (si,1, si,2, ..., si,N)
are equal to the perturbed state si. Those states will be
taken into account similarly to the states different from si.
We then simulate synchronously all the states in (si,1, si,2,
..., si,N) until they reach one of the attractors of the system
(i.e. an element of S). The transition probability from a
cycle to another is defined as the ratio of the number of
perturbed states of the first cycle that reach the second one
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 8 of 14
(page number not for citation purposes)
over the total number of perturbed states of the first cycle.
We also take into account the transitions from a cycle to
itself. By this mean, we add some asynchronous dynamics
in our synchronous analysis. We can then compute the
stationary distribution of this Markov process and inter-
pret it as a measure of the relative importance of each
cycle. We achieve this computation with an initial vector
of size n and of value [1/n, 1/n, ..., 1/n] where n is the size
of S. By this mean, we make sure that all the absorbing
states are taken into consideration. The aim of this meth-
odology is to provide a measure of the stability of each
synchronous attractor, using an asynchronous perturba-
tion. Expecting that all the resulting attractors of synchro-
nous Boolean analysis are biologically relevant would
mean that all the biochemical reactions in the network
happened simultaneously. Yet we know that it is not the
case. Furthermore, it is well known that, within a biologi-
cal pathway, some reactions are triggered with higher fre-
quency than others (that is why, in the ODE paradigm, a
lot of systems are stiff). Thus, Boolean analysis should, in
some way, take into consideration the asynchronous char-
acter of biochemical reactions. Under this assumption,
asynchronous perturbations seem a logical and conven-
ient way to provide a hint of the biological relevance of
the state cycles found.
Results
Dynamical synchronous analysis of the model
Based on the simulations obtained from 105 random
states (out of 233 . 8.6 × 109 possible states), we predict 3
equilibria or steady states:
• one with the complex SREBP-SCAP-Insig1 activated but
a lack of precursor (Acetyl-CoA) preventing cholesterol
synthesis;
• one with the presence of cholesterol precursor (Acetyl-
CoA), but also the presence of statins blocking the choles-
terol synthesis by inhibiting the HMG-CoA reductase
enzyme;
• and one with a lack of precursor and the presence of stat-
ins.
Furthermore, we found 4 state cycles corresponding to the
physiological regulation of cholesterol synthesis: when
the cholesterol level is too low (equivalent to the absence
of cholesterol in a Boolean formalism) there is activation
of the SREBP-SCAP complex and (enhancement of the)
production of all the enzymes of the cholesterol synthesis
regulated by SREBP. Then, the endogenous synthesis of
cholesterol starts again and when its level becomes too
high (equivalent to the presence of cholesterol in a
Boolean formalism) it inhibits the release of the SREBP-
SCAP complex and thus the production of the above
enzymes.
Among those 4 cycles one has size 29 (named cycle_0 fur-
ther in this article) and the others have size 33. In the cycle
of size 29 the cholesterol changes from false to true (i.e.
the cholesterol gets above the threshold indicative of the
activation of its synthesis by the complex SREBP-SCAP-
Insig) only once per cycle, while in the cycles of size 33,
the cholesterol becomes true 5 times per cycle.
Results verification through a formal analysis using a SAT
solver
The results detailed in the previous paragraph are
obtained using a start space for the simulation around 105
times smaller than the state space. This method has the
advantage to quickly provide some attractors for the bio-
logical system. However, when using a sample of the
whole state space, there is no assurance of finding all the
system attractors. Formal analysis is one way to ensure
that all the attractors have been found with a computa-
tional cost that could be lower than the cost of performing
simulation on the whole state space. We decided to per-
form such a formal analysis by running a SAT solver on
our Boolean network. We recall here that the Boolean sat-
isfiability problem (commonly called SAT-problem) [32]
determines if there is a set of variables for which a given
Boolean formula can be evaluated to TRUE and identifies
this precise set if existing. This is an NP-complete problem
for which some instance solvers have been developed. To
achieve this formal analysis, we wrote our system of
Boolean equations into a suitable dimacs file format [33]
(some dimacs files used for simulation can be down-
loaded at http://Bioinformaticsu613.free.fr). In that way,
we were able to confirm that the only attractors of size 1,
29 and 33 were those detected by our simulation tool with
a random start space of size 105.
Why did we need to go further: detection of spurious cycles
The simulation performed with our model results in 4
state cycles. We believe that all those cycles do not corre-
spond to a phenotype. These outcomes of different simu-
lations, which are not biologically relevant, are typical of
the synchronous Boolean paradigm, and are called spuri-
ous cycles [4,5]. Therefore, there is a need to measure the
relative importance of the cycles found using the previous
methods.
Markov chains-based stability analysis of the previous
synchronous simulation
Let us use our stability analysis on the results of the syn-
chronous simulation of the cholesterol regulatory path-
way. We perturb the cyclic attractors found during this
simulation and then simulate synchronously the states
resulting from the perturbation. We interpret the ratio of
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 9 of 14
(page number not for citation purposes)
the number of perturbed states of a given cyclic attractor
that reach a second cyclic attractor over the total number
of perturbed states of the given cyclic attractor as a transi-
tion probability. Afterwards, we use all the computed
transition probabilities obtained previously to build the
Markov chain shown in figure 5.
The stationary probability vector is [1, 0, 0, 0]. This reflects
the fact that the state cycle 0 is absorbing (i.e. it has no
outgoing transitions). We interpret this result as state
cycles 1, 2 and 3 are spurious.
Markov chains-based stability analysis of simple regulatory
networks
Furthermore, to validate our method of Markov chains-
based stability analysis, we applied it on simple positive
and negative regulatory loops of different sizes, which are
well known to contain spurious cycles. In the example of
a negative feedback loop of size 3, the state cycle {010,
101} is found in synchronous simulation but does not
exist in asynchronous simulation. It is obviously spurious,
as detected by our method. All the detailed results and
graphs can be found on our web site: http://
bioinformaticsu613.free.fr/simpleloopsn3.html.
Markov chain of the transition probabilities between state cycles in the cholesterol regulatory pathway
Figure 5
Markov chain of the transition probabilities between state cycles in the cholesterol regulatory pathway. Let k be
the number of states within an attractor (k can be 1 in the case of a point attractor) and N be the number of species in the
model. For each attractor of this finite time-homogeneous Markov chain, we perturb each species of each state by triggering its
own Boolean function asynchronously. Thus there are kN perturbations per attractor. In the cholesterol regulatory pathway,
one cyclic attractor found by the synchronous analysis has 29 states and the three other cyclic attractors have 33 states. The
number of species in the model is 33. The weight of the edge from an attractor X to an attractor Y is the ratio between the
number of perturbations of X which lead to Y over the total number of perturbations of X.
Cycle 0
Cycle 1
Cycle 2
Cycle 3
957/957
33/1089
78/1089
52/1089
33/1089
33/1089
26/1089
1030/1089
978/1089
971/1089
33/1089
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 10 of 14
(page number not for citation purposes)
When we perform this analysis on our example of a sim-
ple regulatory network (figures 1, 2 and 3) we obtain the
results shown in table 2. The transition probabilities asso-
ciated to the simple regulatory network shown in table 1
allow us to build the Markov chain shown in figure 6. The
resulting stationary distribution of the Markov chain
shown in figure 6 is [1, 0]. Thus we interpret the state cycle
of our simple regulatory network as spurious.
Benchmarking of our method
The lack of a common file format for qualitative analysis
is an important issue for the benchmark of such methods.
This is clearly stated in the article about the SQUAD soft-
ware by Di Cara et al [34]: "With the increase of published
signaling networks, it will be possible in the future to realize a
benchmark among these software packages to compare their
strengths and weaknesses. For doing that, however, it would be
very useful to develop a common file format." Since Di Cara's
article, this issue of a common file format for qualitative
analysis is still important because the current SBML for-
mat cannot encode logical models. However, work is
ongoing to extend SBML such that version 3 could sup-
port information for qualitative simulation. The solution
we have found to overcome this current limitation was to
use a proprietary annotation with a specific namespace to
remain compatible with the SBML standard. This allowed
us to read and write annotated SBML files from our com-
putational tool. However, other software cannot use our
annotated SBML to perform the qualitative analysis of a
biological network, unless specific code is developed to
read our annotations. SQUAD can generate SBML, but it
cannot use its own generated SBML to perform qualitative
analysis. All the information concerning qualitative anal-
ysis is stored only in MML files (the file format used by
SQUAD). This is why we could not use the current SBML
version to perform the benchmark.
Furthermore, except for some well-known problems
which have been well formulated and thus accepted by
the community (like the test for Initial Value Problems
(IVPs) solvers of the Bari University [35] or the ISCAS89
benchmark for circuits [36]), any type of benchmark
would be partial and its results could be seen as unfair by
other authors. For example, as mentioned by Naldi et al.
in [37], some methods are only suited for a subset of bio-
logical problems: "Garg et al. have already represented
Boolean state transition graphs in terms of BDD. They consid-
ered the particular case of networks where genes are expressed
provided all their inhibitors are absent and at least one of their
activators is present " [7]. The Naldi et al.'s method, regard-
less of its computational efficiency, is a more powerful
modeling tool thanks to the use of logical evolution rules
and multi-valued species. Even if it is always possible to
use Boolean formalism to model multi-valued networks
(by leveraging the number of Boolean species by the
number
of
wished
values,
e.g.
{Boolean_species_A_low_level,
Boolean_species_A_middle_level,
Boolean_species_A_high_level}), the use of multi-valued
logical networks greatly eases the modeling process.
However, despite all the restrictions discussed above, we
believe that benchmarking our method is an important
issue. We have then developed a program that generates a
random network whose size is a user input. For the sake
of simplicity, the obtained network contains species that
can have 0, 1, 2 or 3 species influencing it. This means
that, to compute the state of a species at time t + 1, we only
Table 2: Computation of the transition probabilities associated to our simple regulatory network.
Attractors
Steady State (SS)
State Cycle (SC)
States
1010
0010
1100
1011
Perturbed states and their limit cycles
1010 → SS
1010 → SS
1100 → SC
0011 → SC
0110 → SS
1000 → SC
1011 → SC
0000 → SC
1110 → SS
1011 → SC
0010 → SC
1101 → SC
1010 → SS
Resulting probability transition
P(SS → SS) = 1
P(SC → SC) = 8/12 . 0.67
P(SC → SS) = 4/12 . 0.33
This table shows both the principle of our newly introduced stability analysis and its application on the simple regulatory network shown in figure 1.
It has 2 main columns: the steady state column and the state cycle column. As the state cycle found during the dynamical synchronous analysis
contains 3 states (see figure 2), the last column is divided into 3 sub-columns. In the line "Perturbed states and their limit cycles" we show the
perturbation results of each state of each attractor by re-evaluating each species by its own Boolean function triggered asynchronously.
Perturbations are done from species A to species D. There are 4 species in our simple regulatory model, therefore 4 new states are generated
from 1 perturbed state. We then synchronously simulate each new state and note if their simulation leads to the attractor they are derived from or
to the other attractor of the system. In other words, we watch in which basin of attraction are those new states (see figure 2). The arrows
following by "SC" (state cycle) or "SS" (steady state) give those responses. For the steady state no perturbation has an effect because a steady state
in synchronous analysis remains a steady state in asynchronous analysis. Thus we simplify the presentation, showing that all the perturbations
applied to state [1010] leave this state unchanged.
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 11 of 14
(page number not for citation purposes)
need to know the state of a maximum of 3 other species at
time t. Our benchmarking tool has 3 parameters that can
modify the connection density of the network :
• the probability of being a "source" (i.e. the probability
for a species to be influenced by no other species in the
network),
• the probability to be under the influence of only one
other species in the network,
• the probability to be under the influence of exactly two
other species in the network.
The complementary probability is the probability for a
species to be influenced by three other species in the net-
work.
Then our benchmarking tool generates the files describing
this network for our software as well as in MML and
GINML formats for SQUAD [34,38] and GINsim [39,40],
respectively.
We have analyzed networks of sizes ranging from 33 (the
number of species in our cholesterol regulatory pathway)
up to 2500 species.
The CPU times obtained with our method on a Intel®
Core™ 2 Duo E6600 processor (2.4 GHz) with 2 GBytes of
RAM are shown in table 3. The performance of other
tested software did not compare favorably with our appli-
cation. With GINsim, we were able to simulate networks
as large as 1000 species, but we obtained an "out of mem-
ory" error message for the network of 2500 species. When
we used the SQUAD software, we were unable to simulate
a network of 1000 species or above. It is however possible
that the parameters used to build the automatically gener-
ated networks might have an impact on the results of the
benchmarking. Nevertheless, under the conditions used,
our application is appropriate for the analysis of large bio-
logical networks.
Discussion
The results reported here are in accordance with the bio-
logical knowledge we have on the cholesterol biosynthe-
sis pathway. The steady states found correspond to either
a lack of precursor (Acetyl-CoA) or arise from the effect of
statins blocking the endogenous synthesis of cholesterol,
Markov chain of the transition probabilities between the steady state and the state cycle in our simple regulatory network
Figure 6
Markov chain of the transition probabilities between the steady state and the state cycle in our simple regula-
tory network. Let k be the number of states within an attractor (k can be 1 in the case of a point attractor) and N be the
number of species in the model. For each attractor of this finite time-homogeneous Markov chain, we perturb each species of
each state by triggering its own Boolean function asynchronously. Thus there are kN perturbations per attractor. In our simple
regulatory network the cyclic attractor has 3 states. The number of species in the model is 4. The weight of the edge from an
attractor X to an attractor Y is the ratio between the number of perturbations of X which lead to Y over the total number of
perturbations of X.
Steady
State
State
Cycle
4/12
12/12
8/12
Table 3: Benchmark of our method for qualitative analysis of biological networks.
number of species
33
100
250
500
1000
2500
CPU time
7.515s
15.874s
39.999s
90.295s
173.107s
569.511s
This table shows the benchmarking results of our qualitative analysis methods on automatically generated networks. Its has been done on a Intel®
Core™ 2 Duo E6600 processor (2.4 GHz) with 2 GBytes of RAM.
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 12 of 14
(page number not for citation purposes)
and the cyclic attractor corresponds to a physiological reg-
ulation of cholesterol synthesis. Based on these data, we
will be able to evaluate the putative impacts of additional
modifications along the pathway. For instance, we may
evaluate the effect of compensatory intermediates such as
farnesyl pyrophosphate or geranylgeranyl pyrophosphate,
which are expected to both restore cholesterol synthesis
and prevent the deleterious effects of its absence [25,41-
44]. These compounds may compensate for the lack of
mevalonate, a condition that could readily be introduced
in our computerized scheme and assayed experimentally
at the same time. This would be particularly relevant in
the field of cancer research, for defects in lipid signalling
are of primary importance [19,45-47]. Hence, secondary
protein modifications, including farnesylation or geranyl
geranylation, which depend on the availability of farnesyl
and geranyl geranyl pyrophosphate, respectively, are
known to play pivotal roles in the progression of tumours
that depend on Ras functional status (for review see [15]).
However, it would require further studies to integrate our
cholesterol regulatory pathway with oncological path-
ways, like the Ras activation pathway. We believe that this
would be a particularly interesting perspective, bearing in
mind that signal transduction pathways with G proteins
have been extensively studied [48-52] and modeling
efforts have already been made [53,54]. Furthermore, the
method described here to identify spurious cycles opens
new routes to compute large and biologically relevant
models thanks to the computational efficiency of syn-
chronous simulation. An important aspect was to bench-
mark our method in order to determine if its
computational efficiency is comparable to those of GIN-
sim and SQUAD. Our results show that our method can
analyze networks containing as many as 2500 species and
was time efficient. Indeed, the approach could well be
applied to other regulatory pathways, either from other
metabolic routes or from transduction signaling. How-
ever, the current model is purely a Boolean model where
a gene is either active or inactive, a protein either present
or not. An obvious limitation of Boolean formalism
comes, for example, from the difficulty or the impossibil-
ity to model a simultaneous and antagonist influence on
a species, e.g. if a gene is under the influence of a silencer
and an activator. In that case, we would like to be able to
model a threshold above which there is activation or inhi-
bition of the targeted species, e.g. there is RNA production
when there is at least twice as much activator as silencer.
Boolean formalism is not suitable for this purpose. This
limitation could however be alleviated by expressing the
presence of a molecular species with an enumeration of
values ranging from the complete lack to a highly over-
expressed level such as in the generalized logical modeling
approach of Thomas and D'Ari [4]. This would also ena-
ble to address, with a more realistic approach, the effect of
an inhibitor or the effect of an enzyme, and to predict the
preponderance of one or the other species in case of antag-
onistic regulation. The multi-level approach was success-
fully applied to many experimentally studied biological
regulatory networks (e.g. [55-58]). We can note here, that
our Markov chains-based stability analysis could readily
be extended on the analysis of a multilevel qualitative
simulation. Other work seems to be ongoing on choles-
terol on cholesterol modeling using a set of ordinary dif-
ferential equations thanks to a huge effort of
identification of biochemical kinetics and this should add
further insights on the understanding of this pathway
[59]. Those two last approaches would allow us, for exam-
ple, to analyze different cholesterol levels.
Conclusion
To the best of our knowledge, this is the first description
of a dynamic systems biology model of the human choles-
terol pathway and several of its key regulatory control ele-
ments. This study was designed with a formal
methodology and was challenged through the use of an
important biochemical pathway. To efficiently analyze
this model and ensure further analysis even after its com-
plexification and possible merge with other pathway
models like Ras signaling cascade models, we associate a
classical and computationally efficient synchronous
Boolean analysis with a newly introduced method based
on Markov chains, which identifies spurious cycles among
the results of the synchronous analysis. The in silico exper-
iments show the blockade of the cholesterol endogenous
synthesis by statins and its regulation by SREPBs, in full
agreement with the known biochemical features of the
pathway. Furthermore, because high throughput experi-
ments give rise to increased complexification of biological
systems, there are major needs for new computational
developments for their dynamical analysis. Our method-
ology is one answer to this new challenge.
Authors' contributions
LC and GK conceived this study and built the model based
on literature data. GK conducted the in silico experiments.
Acknowledgements
We wish to thank very much Dr. Claudine Chaouiya for her critical reading
of the manuscript.
This work was supported by grants from the Inserm, the Brittany region,
the University of Brest and the Medical Faculty of Brest. Gwenael Kervizic
was supported by a CIFRE contract from the ANRT.
References
1.
Kitano H: Computational systems biology. Nature 2002,
420(6912):206-10.
2.
Kauffman SA: Metabolic stability and epigenesis in randomly
constructed genetic nets. J Theor Biol 1969, 22(3):437-67.
3.
Kauffman S: The Origins of Order: Self-organization and Selection in Evolu-
tion Oxford University Press, NY; 1993.
4.
Thomas R, D'Ari R: Biological Feedback CRC Press; 1990.
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 13 of 14
(page number not for citation purposes)
5.
Faure A, Naldi A, Chaouiya C, Thieffry D: Dynamical analysis of a
generic Boolean model for the control of the mammalian
cell cycle. Bioinformatics 2006, 22(14):e124-31.
6.
Chaves M, Albert R, D SE: Robustness and fragility of Boolean
models for genetic regulatory networks. J Theor Biol 2005,
235(3):431-49.
7.
Garg A, Xenarios I, Mendoza L, DeMicheli G: An Efficient method
for dynamic analysis of gene regulatory networks. RECOMB
2007; Heidelberg 2007:62-76.
8.
Horton JD, Shimomura I, Brown MS, Hammer RE, Goldstein JL, Shi-
mano H: Activation of cholesterol synthesis in preference to
fatty acid synthesis in liver and adipose tissue of transgenic
mice overproducing sterol regulatory element-binding pro-
tein-2. J Clin Invest 1998, 101(11):2331-9.
9.
Osborne TF: Sterol regulatory element-binding proteins
(SREBPs): key regulators of nutritional homeostasis and
insulin action. J Biol Chem 2000, 275(42):32379-82.
10.
Shimano H: Sterol regulatory element-binding protein family
as global regulators of lipid synthetic genes in energy metab-
olism. Vitam Horm 2002, 65:167-94.
11.
Auer J, Eber B: A clinical focus on statins. Curr Opin Investig Drugs
2001, 2(3):382-388.
12.
Bocan TM: Pleiotropic effects of HMG-CoA reductase inhibi-
tors. Curr Opin Investig Drugs 2002, 3(9):1312-1317.
13.
Buhaescu I, Izzedine H: Mevalonate pathway: a review of clinical
and therapeutical implications. Clin Biochem 2007, 40(9–
10):575-584.
14.
Fears R: Mode of action of lipid-lowering drugs. Baillieres Clin
Endocrinol Metab 1987, 1(3):727-754.
15.
Demierre MF, Higgins PD, Gruber SB, Hawk E, Lippman SM: Statins
and cancer prevention. Nat Rev Cancer 2005, 5(12):930-42.
16.
Coleman PS, Chen LC, Sepp-Lorenzino L: Cholesterol metabo-
lism and tumor cell proliferation. Subcell Biochem 1997, 28(363–
435):363.
17.
Cox AD, Der CJ: The ras/cholesterol connection: implications
for ras oncogenicity. Crit Rev Oncog 1992, 3(4):365-400.
18.
Edwards PA, Ericsson J: Signaling molecules derived from the
cholesterol biosynthetic pathway: mechanisms of action and
possible roles in human disease. Curr Opin Lipidol 1998,
9(5):433-440.
19.
Rao KN: The significance of the cholesterol biosynthetic path-
way in cell growth and Carcinogenesis. Anticancer Res 1995,
2(2):309-314.
20.
Graaf MR, Richel DJ, van Noorden CJ, Guchelaar HJ: Effects of stat-
ins and farnesyltransferase inhibitors on the development
and progression of cancer. Cancer Treat Rev 2004, 30(7):609-41.
21.
Chan KK, Oza AM, Siu LL: The statins as anticancer agents. Clin
Cancer Res 2003, 9:10-19.
22.
Wong WW, Dimitroulakos J, Minden MD, Penn LZ: HMG-CoA
reductase inhibitors and the malignant cell: the statin family
of drugs as triggers of tumor-specific apoptosis. Leukemia
2002, 16(4):508-519.
23.
Berg J, Tymoczko J, Stryer L: Biochemistry 5th edition. W.H. Freeman
and Company; 2001.
24.
Bengoechea-Alonso MT, Ericsson J: SREBP in signal transduc-
tion: cholesterol metabolism and beyond. Curr Opin Cell Biol
2007, 19(2):215-222.
25.
Brown MS, Goldstein JL: Multivalent feedback regulation of
HMG CoA reductase, a control mechanism coordinating iso-
prenoid synthesis and cell growth. J Lipid Res 1980,
21(5):505-517.
26.
Hucka M, Finney A, Sauro HM, Bolouri H, Doyle JC, Kitano H, Arkin
AP, Bornstein BJ, Bray D, Cornish-Bowden A, Cuellar AA, Dronov S,
Gilles ED, Ginkel M, Gor V, Goryanin I, Hedley WJ, Hodgman TC,
Hofmeyr JH, Hunter PJ, Juty NS, Kasberger JL, Kremling A, Kummer
U, Le Novere N, Loew LM, Lucio D, Mendes P, Minch E, Mjolsness
ED, Nakayama Y, Nelson MR, Nielsen PF, Sakurada T, Schaff JC, Sha-
piro BE, Shimizu TS, Spence HD, Stelling J, Takahashi K, Tomita M,
Wagner J, Wang J: The systems biology markup language
(SBML): a medium for representation and exchange of bio-
chemical network models. Bioinformatics 2003, 19(4):524-31.
27.
Finney A, Hucka M: Systems biology markup language: Level 2
and beyond. Biochem Soc Trans 2003, 31(Pt 6):1472-1473.
28.
Le Novere N, Bornstein B, Broicher A, Courtot M, Donizelli M,
Dharuri H, Li L, Sauro H, Schilstra M, Shapiro B, Snoep JL, Hucka M:
BioModels Database: a free, centralized database of curated,
published, quantitative kinetic models of biochemical and
cellular systems. Nucleic Acids Res 2006:D689-91.
29.
Choudhary A, Datta A, Bittner ML, Dougherty ER: Intervention in
a family of Boolean networks. Bioinformatics 2006, 22(2):226-32.
30.
Shmulevich I, Dougherty ER, Kim S, Zhang W: Probabilistic
Boolean Networks: a rule-based uncertainty model for gene
regulatory networks. Bioinformatics 2002, 18(2):261-74.
31.
Chen PC, Chen JW: A Markovian approach to the control of
genetic regulatory networks. Biosystems 2007, 90(2):535-45.
32.
Du D, Gu J, Pardalos PM: Satisfiability Problem: Theory and Applications
AMS, DIMACS; 1997.
33.
Satisfiability Suggested Format [http://www.satlib.org/Bench
marks/SAT/satformat.ps]. To read this reference you will need the 2
following software: GPL Ghostscript and GSview. GPL Ghostscript
can be downloaded from: http://pages.cs.wisc.edu/~ghost/doc/GPL/
gpl863.htm and GSview can be downloaded from: http://
pages.cs.wisc.edu/~ghost/gsview/get49.htm
34.
Di Cara A, Garg A, De Micheli G, Xenarios I, Mendoza L: Dynamic
simulation of regulatory networks using SQUAD. BMC Bioin-
formatics 2007, 8(462):.
35.
Web page of test for Initial Value Problems (IVPs) solvers
[http://www.dm.uniba.it/~testset]
36.
Web page describing the ISCAS89 benchmark circuits
[http://courses.ece.uiuc.edu/ece543/iscas89.html]
37.
Naldi A, Thieffry D, Chaouiya C: Decision Diagrams for the Rep-
resentation and Analysis of Logical Models of Genetic Net-
works. In Computational Methods in Systems Biology, CMSB 2007,
Edinburgh, Scotland Volume 4695. Springer; 2007:233-247.
38.
Mendoza L, Xenarios I: A method for the generation of stand-
ardized qualitative dynamical systems of regulatory net-
works. Theor Biol Med Model 2006, 3:13.
39.
Chaouiya C, Remy E, Mossé B, Thieffry D: Qualitative analysis of
regulatory graphs: a computational tool based on a discrete
formal framework. In "First Multidisciplinary International Symposium
on Positive Systems: Theory and Applications" (POSTA 2003), of LNCIS Vol-
ume 294. Springer-Verlag; 2003:119-126.
40.
Gonzalez A, Naldi A, Sãnchez L, Thieffry D, Chaouiy C: GINsim : a
software suite for the qualitative modelling, simulation and
analysis of regulatory networks. Biosystems 2006, 84(2):91-100.
41.
Espenshade PJ, Hughes AL: Regulation of sterol synthesis in
eukaryotes. Annu Rev Genet 2007, 41:407-427.
42.
Goldstein JL, Brown MS: Regulation of the mevalonate pathway.
Nature 1990, 343(6257):425-430.
43.
Ness GC, Chambers CM: Feedback and hormonal regulation of
hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase:
the concept of cholesterol buffering capacity. Proc Soc Exp Biol
Med 2000, 224:8-19.
44.
Osborne TF: Transcriptional control mechanisms in the regu-
lation of cholesterol balance. Crit Rev Eukaryot Gene Expr 1995,
5(3–4):317-335.
45.
Chen YQ, Edwards IJ, Kridel SJ, Thornburg T, Berquin IM: Dietary
fat-gene interactions in cancer. Cancer Metastasis Rev 2007,
26(3–4):535-551.
46.
Logette E, Le Jossic-Corcos C, Masson D, Solier S, Sequeira-Legrand
A, Dugail I, Lemaire-Ewing S, Desoche L, Solary E, Corcos L: Cas-
pase-2, a novel lipid sensor under the control of sterol regu-
latory element binding protein 2. Mol Cell Biol 2005,
25(21):9621-9631.
47.
Logette E, Solary E, Corcos L: Identification of a functional DNA
binding site for the SREBP-1c transcription factor in the first
intron of the human caspase-2 gene. Biochim Biophys Acta 2005,
1738(1–3):1-5.
48.
Kamimura D, Ishihara K, Hirano T: IL-6 signal transduction and
its physiological roles: the signal orchestration model. Rev
Physiol Biochem Pharmacol 2003, 149:1-38.
49.
Kandpal RP: Rho GTPase activating proteins in cancer pheno-
types. Curr Protein Pept Sci 2006, 7(4):355-65.
50.
Fritz G, Kaina B: Rho GTPases: promising cellular targets for
novel anticancer drugs. Curr Cancer Drug Targets 2006, 6:1-14.
51.
Bustelo XR, Sauzeau V, Berenjeno IM: GTP-binding proteins of
the Rho/Rac family: regulation, effectors and functions in
vivo. Bioessays 2007, 29(4):356-70.
52.
Strange PG: Signaling mechanisms of GPCR ligands. Curr Opin
Drug Discov Devel 2008, 11(2):196-202.
Publish with BioMed Central and every
scientist can read your work free of charge
"BioMed Central will be the most significant development for
disseminating the results of biomedical research in our lifetime."
Sir Paul Nurse, Cancer Research UK
Your research papers will be:
available free of charge to the entire biomedical community
peer reviewed and published immediately upon acceptance
cited in PubMed and archived on PubMed Central
yours — you keep the copyright
Submit your manuscript here:
http://www.biomedcentral.com/info/publishing_adv.asp
BioMedcentral
BMC Systems Biology 2008, 2:99
http://www.biomedcentral.com/1752-0509/2/99
Page 14 of 14
(page number not for citation purposes)
53.
Singh A, Jayaraman A, Hahn J: Modeling regulatory mechanisms
in IL-6 signal transduction in hepatocytes. Biotechnol Bioeng
2006, 95(5):850-62.
54.
Maurya MR, Bornheimer SJ, Venkatasubramanian V, Subramaniam S:
Reduced-order modelling of biochemical networks: applica-
tion to the GTPase-cycle signalling module. Syst Biol (Steve-
nage) 2005, 152(4):229-242.
55.
Thomas R, Thieffry D, Kaufman M: Dynamical behaviour of bio-
logical regulatory networks-I. Biological role of feedback
loops and practical use of the concept of the loop-character-
istic state. Bull Math Biol 1995, 57(2):247-76.
56.
Sanchez L, van Helden J, Thieffry D: Establishement of the dorso-
ventral pattern during embryonic development of dro-
sophila melanogasater: a logical analysis. J Theor Biol 1997,
189(4):377-89.
57.
Sanchez L, Thieffry D: A logical analysis of the Drosophila gap-
gene system. J Theor Biol 2001, 211(2):115-41.
58.
Schaub MA, Henzinger TA, Fisher J: Qualitative networks: a sym-
bolic approach to analyze biological signaling networks. BMC
Syst Biol 2007, 1:4.
59.
Ratushny A, Ignatieva E, Matushkin Y, Likhoshvai V: Mathematical
model of cholesterol biosynthesis regulation in the cell. Pro-
ceedings of the Second International Conference on Bioinformatics of
Genome Regulation and Structure: 7–11 August 2000; Novosibirsk, Russia
2000:203-06.
60.
CellDesigner [http://www.celldesigner.org/]
61.
Kitano H, Funahashi A, Matsuoka Y, Oda K: Using process dia-
grams for the graphical representation of biological net-
works. Nat Biotechnol 2005, 23(8):961-6.
|
19025648
|
SREBP_SCAP = ( ( Insig_SREBP_SCAP ) AND NOT ( Statins ) )
Mevalonyl_pyrophosphate = ( Mevalonic_Acid )
mSREBP = ( pSREBP )
Farnesyl_pyrophosphate = ( ( Geranyl_pyrophosphate ) AND NOT ( FPP_Synthase ) )
Desmosterol = ( Lanosterol )
Acetyl_CoA_acetyltransferase = ( Acetyl_CoA_acetyltransferase_RNA )
HMG_CoA_Synthase_gene = ( mSREBP )
FPP_Synthase_gene = ( mSREBP )
FPP_Synthase = ( FPP_Synthase_RNA )
Statins = ( Statins )
Cyp51_RNA = ( Cyp51_gene )
Squaline = ( Farnesyl_pyrophosphate )
HMG_CoA_Synthase_RNA = ( HMG_CoA_Synthase_gene )
Dimethyl_allyl_pyrophosphate = ( Isopentenyl_pyrophosphate )
Insig_SREBP_SCAP = NOT ( ( Cholesterol ) )
HMG_CoA_Reductase = ( ( HMG_CoA_Reductase_RNA ) AND NOT ( Statins ) )
Isopentenyl_pyrophosphate = ( Mevalonyl_pyrophosphate )
pSREBP = ( SREBP_SCAP )
HMG_CoA_Synthase = ( HMG_CoA_Synthase_RNA )
Septdehydrocholesterol = ( Lanosterol )
FPP_Synthase_RNA = ( FPP_Synthase_gene )
HMG_CoA_Reductase_gene = ( mSREBP )
Geranyl_pyrophosphate = ( Dimethyl_allyl_pyrophosphate ) OR ( Isopentenyl_pyrophosphate )
HMG_CoA = ( Acetoacetyl_CoA AND ( ( ( HMG_CoA_Synthase AND Acetyl_CoA ) ) ) )
Mevalonic_Acid = ( HMG_CoA AND ( ( ( HMG_CoA_Reductase ) ) ) )
Acetyl_CoA_acetyltransferase_gene = ( mSREBP )
Acetyl_CoA = ( Acetyl_CoA )
Lanosterol = ( Squaline )
Cyp51_gene = ( mSREBP )
HMG_CoA_Reductase_RNA = ( HMG_CoA_Reductase_gene )
Cholesterol = ( Septdehydrocholesterol ) OR ( Desmosterol )
Acetoacetyl_CoA = ( Acetyl_CoA AND ( ( ( Acetyl_CoA_acetyltransferase ) ) ) )
Acetyl_CoA_acetyltransferase_RNA = ( Acetyl_CoA_acetyltransferase_gene )
Cyp51 = ( Cyp51_RNA )
|
BioMed Central
Page 1 of 20
(page number not for citation purposes)
BMC Systems Biology
Open Access
Research article
Modeling ERBB receptor-regulated G1/S transition to find novel
targets for de novo trastuzumab resistance
Özgür Sahin*1, Holger Fröhlich1, Christian Löbke1,2, Ulrike Korf1,
Sara Burmester1, Meher Majety1,3, Jens Mattern1, Ingo Schupp1,
Claudine Chaouiya4, Denis Thieffry4, Annemarie Poustka1,
Stefan Wiemann1, Tim Beissbarth1 and Dorit Arlt*1
Address: 1Division of Molecular Genome Analysis, German Cancer Research Center, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany,
2Current address: Phadia GmbH, Munzinger Strasse 7, 79010 Freiburg, Germany, 3Current address: Roche Diagnostics GmbH, Nonnenwald 2,
82377 Penzberg, Germany and 4Technologies Avancées pour le Génome et la Clinique, INSERM U928, Université de la Méditerranée, Campus
Scientifique de Luminy – Case 928, 13288 Marseille, France
Email: Özgür Sahin* - oe.sahin@dkfz-heidelberg.de; Holger Fröhlich - h.froehlich@dkfz-heidelberg.de;
Christian Löbke - christian.loebke@phadia.com; Ulrike Korf - u.korf@dkfz-heidelberg.de; Sara Burmester - s.burmester@dkfz-heidelberg.de;
Meher Majety - meher.majety@roche.com; Jens Mattern - jens.mattern@dkfz-heidelberg.de; Ingo Schupp - i.schupp@dkfz-heidelberg.de;
Claudine Chaouiya - claudine.chaouiya@univmed.fr; Denis Thieffry - thieffry@tagc.univ-mrs.fr; Annemarie Poustka - a.poustka@dkfz-
heidelberg.de; Stefan Wiemann - s.wiemann@dkfz-heidelberg.de; Tim Beissbarth - t.beissbarth@dkfz-heidelberg.de; Dorit Arlt* - d.arlt@dkfz-
heidelberg.de
* Corresponding authors
Abstract
Background: In breast cancer, overexpression of the transmembrane tyrosine kinase ERBB2 is an adverse prognostic
marker, and occurs in almost 30% of the patients. For therapeutic intervention, ERBB2 is targeted by monoclonal
antibody trastuzumab in adjuvant settings; however, de novo resistance to this antibody is still a serious issue, requiring
the identification of additional targets to overcome resistance. In this study, we have combined computational
simulations, experimental testing of simulation results, and finally reverse engineering of a protein interaction network
to define potential therapeutic strategies for de novo trastuzumab resistant breast cancer.
Results: First, we employed Boolean logic to model regulatory interactions and simulated single and multiple protein
loss-of-functions. Then, our simulation results were tested experimentally by producing single and double knockdowns
of the network components and measuring their effects on G1/S transition during cell cycle progression. Combinatorial
targeting of ERBB2 and EGFR did not affect the response to trastuzumab in de novo resistant cells, which might be due
to decoupling of receptor activation and cell cycle progression. Furthermore, examination of c-MYC in resistant as well
as in sensitive cell lines, using a specific chemical inhibitor of c-MYC (alone or in combination with trastuzumab),
demonstrated that both trastuzumab sensitive and resistant cells responded to c-MYC perturbation.
Conclusion: In this study, we connected ERBB signaling with G1/S transition of the cell cycle via two major cell signaling
pathways and two key transcription factors, to model an interaction network that allows for the identification of novel
targets in the treatment of trastuzumab resistant breast cancer. Applying this new strategy, we found that, in contrast to
trastuzumab sensitive breast cancer cells, combinatorial targeting of ERBB receptors or of key signaling intermediates
does not have potential for treatment of de novo trastuzumab resistant cells. Instead, c-MYC was identified as a novel
potential target protein in breast cancer cells.
Published: 1 January 2009
BMC Systems Biology 2009, 3:1
doi:10.1186/1752-0509-3-1
Received: 9 September 2008
Accepted: 1 January 2009
This article is available from: http://www.biomedcentral.com/1752-0509/3/1
© 2009 Sahin et al; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 2 of 20
(page number not for citation purposes)
Background
Anticancer drugs which are in clinical use show their effect
by acting as non-selective anti-proliferative agents which
kill also the proliferating normal cells in the tumor micro-
environment [1]. The past few decades witnessed the
development of targeted therapies including monoclonal
antibodies, which aim at targeting certain antigens
expressed on the surface of cancer cells with high specifi-
city. In particular, adding trastuzumab, a recombinant
humanized monoclonal antibody directed against the
ectodomain of the receptor tyrosine kinase ERBB2, to reg-
imens containing existing chemotherapeutic agents has
significantly improved clinical outcomes for breast cancer
patients. However, de novo and acquired resistance to tar-
geted therapeutics are common and the next challenges
for the contemporary cancer researchers [2].
The ERBB family of receptor tyrosine kinases is composed
of four receptors that have the ability to form homo- and
heterodimers, and couple binding of extracellular growth
factors to intracellular signal transduction pathways [3,4].
ERBB2, the main player of the ERBB network, does not
show any ligand binding activity, but has high dimeriza-
tion affinity [5,6]. The abnormal activation of ERBB recep-
tors through gene amplification, mutations, or protein
overexpression has been linked to breast cancer prognosis
[7]. Trastuzumab is administrated to ERBB2-overexpress-
ing breast cancer patients [8,9]. The drug shows its effect
by inducing antibody-dependent cellular cytotoxicity
(ADCC), disrupting the downstream signaling of ERBB2
and also resulting in G1/S cell cycle arrest [10]. However,
the response rate to trastuzumab is rather low, with a
range from 12% to 34% having been reported for a
median duration of 9 months [11,12]. Hence, at least two
third of the patients are de novo resistant. On the cellular
level, this might be caused by cancer cells being able to
overcome cell cycle arrest despite targeting the ERBB2
receptor. Therefore, additional targets have to be identi-
fied, which should avoid bypass of cell cycle arrest mech-
anisms.
The cell cycle of eukaryotic organisms is tightly regulated
by the cyclin-dependent kinases (CDKs) and their activa-
tion partners, cyclins [13], which lead cells through the
well-ordered G1-, S-, G2-, and M-phases. It has been
shown that ERBB2 regulates G1/S transition during cell
cycle progression by modulating the activity of the Cyclin
D, Cyclin E/CDK complex, the c-MYC oncogene, and the
p27 kinase inhibitor [7,14]. The restriction points within
different cell cycle phases represent key checkpoints,
where the critical decisions are made for the cells to
divide. At the G1/S restriction point of the cell cycle, cells
are committed to enter S phase where DNA replication
takes place [15]. This process is regulated by Cyclin D/
CDK4/6 and Cyclin E/CDK2 complexes, which phospho-
rylate and thereby inactivate tumor suppressor retinoblas-
toma protein pRB [16-18]. Hyperphosphorylation of pRB
results in the release of the E2F transcription factor that
then initiates the transcription of essential genes for DNA
replication [19]. In both normal and tumor cells, pRB
oscillates between an active (hypophosphorylated) state
in early G1 and an inactive (hyperphosphorylated) state
in the late G1, S and G2/M phases [18]. Therefore, phos-
phorylation and subsequent inactivation of pRB repre-
sents a key event governing cell proliferation.
There have been few studies which applied systems biol-
ogy approaches to identify novel markers [20] and to
define drug target networks in human cancer and other
pathologies [21]. In this study, we focused on the regula-
tion of pRB through ERBB-receptor signaling at a network
level in a de novo trastuzumab resistant cell system to iden-
tify new potential perturbation points leading to cell cycle
arrest. Instead of single candidate gene approach, which
generally examines the role of a single protein considering
it either in conjunction with a second protein or regard-
less to other proteins, we applied a systems biology
approach to identify the role of each component in the
context of protein interaction networks. This strategy is
motivated by the fact that cells react to perturbation of a
single protein by taking advantage of using alternative
ways to keep the system robust. In drug resistance, these
alternative ways allow bypassing the inhibitory effect of
drug treatment. Therefore, in order to find the uncommon
perturbations to which cells cannot find an efficient way
to react, we first integrated published data to build the
protein network for ERBB-receptor regulated cell cycle
progression, then combined qualitative dynamical mode-
ling and robust experimental approaches, and finally pre-
dicted suitable and efficient targets for individual or
combinatorial treatments in de novo trastuzumab resist-
ance in breast cancer as a model system.
We used the Boolean logical framework for the dynamical
modeling and analysis of the biological network. This
framework simplifies the regulatory activity of proteins by
considering them as all or none devices. More precisely,
each protein is defined as being either active (value 1) or
inactive (value 0) depending on its abundance or activity
level. We selected 18 proteins connecting ERBB receptor
signaling to the G1/S transition of cell cycle, and defined
logical rules to describe their regulations with regard to lit-
erature information. Modeling and loss of function simu-
lations of the network proteins were performed using the
modeling and simulation software GINsim [22,23]http://
gin.univ-mrs.fr/GINsim.
Experimental perturbations of each network element
using RNAi and following measurements of their effects
on the output protein allowed us to compare simulations
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 3 of 20
(page number not for citation purposes)
with the experimental results. Quantitative measurements
of protein abundance and activation states using reverse
phase protein arrays (RPPA) [24,25] enabled us to reverse
engineer the interactions of proteins in the cell system we
used, and to compare the experimental network data with
published results of single protein analysis. Utilizing spe-
cific inhibitors against potential targets alone or in combi-
nation with trastuzumab, we further validated the RNAi
experiments and finally defined potential future therapeu-
tic strategies.
Results
Characterization of the de novo trastuzumab resistant cell
system
We first identified a suitable de novo trastuzumab resistant
cell system as prerequisite for studying the ERBB-receptor
regulated network. This cell system should have high
ERBB2 expression but be resistant to trastuzumab treat-
ment. To this end, we first analyzed several breast carci-
noma cell lines and the normal epithelial MCF-12A cell
line for their expression of ERBB family receptors at
mRNA and protein levels, respectively (Figure 1A and 1B).
HCC1954 cells, like SK-BR-3 and BT474 cells, express
high levels of EGFR (ERBB1) and ERBB2 receptors, but
have low levels of ERBB3. ERBB4 receptor expression was
not detected in the HCC1954 cell line.
Next, we examined the response to trastuzumab treatment
of breast carcinoma cells with high ERBB2 expression
(HCC1954 and SK-BR-3) as compared to cells with low
ERBB2 level (MCF-7) in a viability assay (Figure 1C). Cells
were treated with or without trastuzumab and cell viabil-
ity was assayed over time to observe the effect of the drug.
We further tested different concentrations of trastuzumab
to rule out the possibility that the resistance of cell system
could have been due to insufficient amounts of trastuzu-
mab being present in the assay (Additional file 1, Figure
1A). While SK-BR-3 cells responded to trastuzumab start-
ing from day two, HCC1954 cells were resistant, as they
did not show any response to the drug over four days.
Lastly, we verified the resistance of HCC1954 cells to tras-
tuzumab in 3-D cell culture (Figure 1D). After eight days
of treatment, HCC1954 cells were still growing in a large
cluster-like structure that was similar to untreated
HCC1954 cells, whereas SK-BR-3 cells were sensitive to
trastuzumab also in 3-D culture. We could exclude that
the resistance phenotype of HCC1954 cells was due to a
higher or lower ERBB2 expression level compared to sen-
sitive SK-BR-3 cells (Additional file 1, Figure 1B). Further-
more, to rule out the potential impact of possible
mutations in the ERBB2 protein on resistance phenotype
of the HCC1954 cells, the ERBB2 gene sequence was veri-
fied by sequencing and no mutation was found. Hence,
HCC1954 cells were chosen as a de novo trastuzumab
resistant model cell system in this study.
Determination of experimental output
Next, we characterized HCC1954 cells with regard to G1/
S progression by measuring the levels of pRB phosphor-
ylation, and of cell cycle proteins by comparing MOCK
(only lipofectamine transfection reagent) and CDK4
siRNA transfected cells. After synchronization, we stimu-
lated the cells with EGF. Starting from "0 hour (no EGF)",
cells were lysed at different time points and proteins of
interest were detected with specific antibodies (Figure
2A). CDK4 knockdown was efficient, as no residual pro-
tein was visible on the blot. Due to Dif-3 treatment, which
degrades Cyclin D1 at both mRNA and protein levels [26],
the level of Cyclin D1 was low at 0 h while it increased
upon continuous EGF stimulation. After 6 h of EGF stim-
ulation, Cyclin D1 expression remained constant until 24
hours in both MOCK-treated cells and after CDK4 knock-
down. For the MOCK control, a gradual increase in the
Cyclin E1 level was observed, starting from EGF stimula-
tion (0 hour) to 18 hours. In contrast, Cyclin E1 expres-
sion did not change from 0 h to 18 h after CDK4
knockdown. Surprisingly, we observed a reduction also of
CDK2 in the CDK4-siRNA treated cells, starting at 6 h.
This might be due to the partnering of CDK2 with Cyclin
E1, whose level did not increase in case of CDK4 knock-
down.
We found the phosphorylation of pRB (Ser 807/811), our
marker for G1/S transition, to be delayed and the pRB
expression level to be decreased after CDK4 knockdown,
as compared to the MOCK control. Hence, we next quan-
tified the phosphorylation level of pRB (Figure 2B) in the
same lysates using reverse phase protein arrays (RPPA).
The phosphorylation level of pRB was found to be low in
the growth-arrested cells and induction of pRB phosphor-
ylation from 6 to 12 hours did not occur abruptly for
CDK4 knockdown compared to MOCK. These data dem-
onstrate that phosphorylation of pRB at the transition
point can be quantified by RPPA as an output of EGF stim-
ulation.
Literature-based Boolean network of G1/S transition
The initial network of G1/S transition was built by extract-
ing information from the literature about interactions
between proteins involved in receptor tyrosine kinase-reg-
ulated cell cycle progression (Additional file 1, Table 1).
The resulting network encompasses 18 proteins, includ-
ing EGF as stimulus, homo- and heterodimers of ERBB
family members, tyrosine kinase receptor IGF-1R, key
transcription factors (ER-α and c-MYC), key signaling
intermediates (AKT1 and MEK1), and G1/S transition cyc-
lins, CDKs and CDK inhibitors (Figure 3). Upon activa-
tion, members of the ERBB family of tyrosine kinases
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 4 of 20
(page number not for citation purposes)
Characterization of breast cell lines for trastuzumab resistance/sensitivity
Figure 1
Characterization of breast cell lines for trastuzumab resistance/sensitivity. A. qRT-PCR to determine ERBB recep-
tor family expression at mRNA level in MCF-12A normal breast epithelial cells and in five breast carcinoma cells. B. Western
blots showing the expression level of ERBB family receptors. HCC1954 cells express high levels of ERBB1 and ERBB2 recep-
tors, but low level of ERBB3 and no ERBB4. β-actin was used as loading control. C. WST-1 cell viability assay to assess the
resistance of breast carcinoma cells to trastuzumab (100 nM). Compared to SK-BR-3 cells, with high level of ERBB2 receptors,
HCC1954 cells are resistant to trastuzumab (100 nM) over 4 days. D. Verification of resistance of HCC1954 cells to trastuzu-
mab compared to sensitive SK-BR-3 cells in 3-dimensional cell culture. Photos were taken after eight days of trastuzumab
treatment.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 5 of 20
(page number not for citation purposes)
form homo- and heterodimers. For HCC1954 cells, six
different such dimers are possible as ERBB4 is not
expressed in this cell line. The dimers ERBB1/ERBB2,
ERBB1/ERBB3 and ERBB2/ERBB3 were represented as
specific nodes in the network. Homodimers were implic-
itly represented by the corresponding protein nodes. Since
no ligand is known for ERBB2 homodimers [27] and as
ERBB3 homodimers have a defective tyrosine kinase
domain [28], the corresponding nodes are unable to acti-
vate the ERBB targets AKT1 and MEK1. The effects of the
combinations of interactions on the activity of each pro-
tein was defined in terms of logical rules using the
Boolean operators AND, OR, and NOT. Table 1 lists these
Boolean rules for the network components, and data sup-
porting the rules is provided in the Additional file 1, Table
1. We then utilized the modeling and simulation software
GINsim [23] to implement these rules into a computa-
tional model. Figure 3 shows the resulting logical regula-
tory graph for the ERBB receptor-regulated G1/S transition
protein network. Normal arrows denote positive regula-
tions, which are either through phosphorylation, tran-
scriptional activation, or physical interaction (e.g,.
complex formation). Blunt-ended arrows denote negative
regulations. The numbers associated with each edge refers
to the respective publications providing experimental
data in support of the corresponding regulatory interac-
tion (Additional file 1, Table 1).
Simulation of loss-of-functions
For loss-of-function simulations, we performed in silico
knockdowns of the network proteins by fixing the level of
the perturbed element to "0", meaning that the corre-
sponding protein was always "inactive" (Additional file 1,
Figure 2). Each simulation was performed for specific ini-
tial protein states, matching the biological and experi-
mental conditions (for several proteins, we considered
both possible values). For example, the initial states of
p21 and p27, both of which being CDK inhibitors, were
set to "1" because, in G0/G1 arrested cells, the expression
levels of these inhibitors are high and their levels decrease
(due to their degradation in proteasomes) once cells
progress through S phase [29]. To represent continuous
EGF simulation, the initial values of ERBB nodes were set
to "1". Since the cells had been synchronized with Dif-3,
which degrades Cyclin D1, the initial level of Cyclin D1
was set to "0". Using the resulting initial states, we com-
puted all possible state transitions and iterated until we
finally obtained a unique "stable state" in which the level
of each protein was fixed (details about knockdown sim-
ulations can be found in Materials and Methods section).
Proof of principle experiments for the determination of G1/S transition point in trastuzumab resistant HCC1954 cells
Figure 2
Proof of principle experiments for the determination of G1/S transition point in trastuzumab resistant
HCC1954 cells. A. Western blots showing the expression and activation of key G1/S proteins. Cells were treated either only
with Lipofectamine (MOCK) or with 20 nM CDK4 siRNA for 24 hours. Then, cells were synchronized for 24 hours and subse-
quently stimulated with 25 ng/ml EGF for 6, 12, 18 or 24 hours. Cell lysates were applied to immunoblotting. B. Reverse Phase
Protein Array (RPPA) showing the phosphorylation state of pRB protein (Ser 807/811). The same lysates (from A) were
applied to RPPA. The upper panel shows the read-out of antibody signal at near infra-red range for phospho-pRB antibody with
four replicates. The lower panel shows the graphical representation of phospho-pRB antibody signal intensity for two different
conditions and for four time points. Signals were normalized to 0 hour MOCK sample.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 6 of 20
(page number not for citation purposes)
Table 2 summarizes the outcomes of the simulations. Out
of 17 loss-of-function simulations, a significant decrease
of pRB phosphorylation (pRB is predominantly in its
hyphophosphorylated form and cells do not progress
through G1/S transition: pRB = 0) was predicted for
CDK4, Cyclin D1, and CDK6 loss of functions. For the
ERBB1_2 and ERBB1_3 knockdowns, we obtained two
possible stable states characterized by pRB = 0 and 1 that
should be resolved with experimental results. The loss-of-
function simulations for all other network proteins
resulted in the preservation of pRB phosphorylation (pRB
= 1), thus tentatively enabling G1/S transition.
Furthermore, we have also simulated the loss-of-function
of multiple proteins (all double and many triple knock-
downs) (Additional file 2, Table 1). We have observed
that if one protein knockdown gives pRB = 0, the combi-
nation of any other protein knockdown with this one also
gives pRB = 0 (e.g. ER-α knockdown gives pRB = 0 and ER-
α+AKT1 knockdown also gives pRB = 0) and we have ver-
ified this experimentally as well (Additional file 2, Table
2). We have also simulated the knockdown of all three
receptors at the same time (ERBB1_2_3) and it resulted in
2 stables states with pRB = 1 or 0 (Additional file 2, Table
1).
Assessment of siRNA knockdown efficiency and specificity
for experimental testing of simulations
In order to validate the simulations having been per-
formed for various possible loss of functions, we utilized
RNAi to experimentally induce knockdown of the corre-
sponding proteins. First, we validated the siRNAs accord-
ing to their knockdown efficiency at both mRNA and
protein levels by qRT-PCR and Western blotting, respec-
tively (Figure 4A and 4B). We obtained at least 70%
knockdown at mRNA level for all the network proteins
(Figure 4A) and for most a similar knockdown also at the
protein level (Figure 4B), both in single and combinato-
rial RNAi settings [24]. Because of the high level of
sequence conservation among ERBB family receptors
[27], it was imperative to test for a potential cross-reactiv-
ity of ERBB receptor siRNAs (Figure 4C and 4D). To this
end, we compared the effects of the pools of four siRNAs
for every gene with those of individual siRNAs. Neither
pools nor individual siRNAs were found to have cross-
reactivity (Figure 4C and 4D).
In the combinatorial RNAi settings, the levels of the ERBB
proteins in the EGFR/ERBB2, EGFR/ERBB3, and ERBB2/
ERBB3 heterodimers were efficiently downregulated (Fig-
ure 4B). While the EGFR level was drastically reduced
when we applied double knockdown of EGFR/ERBB2,
EGFR/ERBB3, and ERBB2/ERBB3 (Figure 4B), the EGFR
protein was stable in the single knockdown with EGFR
siRNA although this treatment resulted in more than 80%
knockdown at the mRNA level (Figure 4A). However,
knockdown of the ERBB2 receptor resulted in a substan-
tial decrease of EGFR at the protein level. With respect to
the qRT-PCR results, we can exclude that this effect could
be due to a cross-reaction of the ERBB2 siRNA (Figure
4C). Therefore, we hypothesize an indispensable partner-
ing of ERBB2 and EGFR in ERBB2 overexpressing cells
(Figure 4D), and assume that the EGFR receptor protein is
efficiently stabilized that way. This hypothesis was sup-
ported by similar observations made in ERBB2 overex-
pressing SK-BR-3 cells, but not in MDA-MB-231 having
low ERBB2 expression (Additional file 1, Figure 3).
Experimental validation of loss-of-function simulations
Next, we designed a series of in vitro experiments using the
validated conditions described above to assess the results
from loss of function simulations. Lysates of three biolog-
ical replicates were analyzed with RPPA using four techni-
cal
replicates
of
each.
The
signal
intensity
of
phosphorylated pRB was measured in the near-infrared
range (NIR) for each knockdown at two time points (0 h
and 12 h). As a negative control, we utilized MOCK sam-
ples which had not been stimulated with EGF. Results
were compared to MOCK samples (reference sample),
which had been stimulated with EGF, and the significance
of the impact on pRB phosphorylation was tested using
Table 1: Boolean rules for the activation of each component of
the network presented in Figure 3.
Target
Logical rules for the activation of target
ERBB1
EGF
ERBB2
EGF
ERBB3
EGF
ERBB1_2
ERBB1 Λ ERBB2
ERBB1_3
ERBB1 Λ ERBB3
ERBB2_3
ERBB2 Λ ERBB3
IGF1R
(ER-α V AKT1) Λ !ERBB2_3
ER-α
AKT1 V MEK1
c-MYC
AKT1 V MEK1 V ER-α
AKT1
ERBB1 V ERBB1_2 V ERBB1_3 V ERBB2_3 V IGF1R
MEK1
ERBB1 V ERBB1_2 V ERBB1_3 V ERBB2_3 V IGF1R
CDK2
Cyclin E1 Λ !p21 Λ !p27
CDK4
Cyclin D1 Λ !p21 Λ !p27
CDK6
Cyclin D1
Cyclin D1
AKT1 V MEK1 V ER-α V c-MYC
Cyclin D1*
ER-α Λ c-MYC Λ (AKT1 V MEK1)
Cyclin E1
c-MYC
p21
ER-α Λ !AKT1 Λ !c-MYC Λ !CDK4
p27
ER-α Λ !CDK4 Λ !CDK2 Λ !AKT1 Λ !c-MYC
pRB
(CDK4 Λ CDK6)V (CDK4 Λ CDK6 Λ CDK2)
These rules were derived from the references listed in the Additional
file 1, Table 1. The refined rules for Cyclin D1 are shown with "*".
Symbols "Λ": AND, "V": OR and "!": NOT
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 7 of 20
(page number not for citation purposes)
ERBB receptor regulated G1/S transition network derived from published data
Figure 3
ERBB receptor regulated G1/S transition network derived from published data. ERBB receptors are functional (i.e.
able to transmit signal to downstream proteins) only when they form heterodimers, except for ERBB1, which is also functional
as a homodimer. The number associated with each arrow indicates the reference from which the corresponding interaction
was extracted (a list of these references is provided in the Additional file 1, Table 1). Normal arrows denote positive regula-
tions, whereas blunt arrows denote negative regulations. These interactions correspond to transcriptional regulations, post-
transcriptional modifications, or physical interaction. EGF constitutes the input and pRB protein represents the output of the
network.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 8 of 20
(page number not for citation purposes)
the ANOVA method. Box plots of the knockdown effect
on pRB phosphorylation are shown in Figure 5A. We clas-
sified effects as "1" in cases where the knockdown of a spe-
cific protein had resulted in a phosphorylation profile
similar to the MOCK profile at 12 h, and no significant
change of the pRB phosphorylation state had been
observed. Conversely, if the knockdown resulted in a sig-
nificantly lower pRB phosphorylation level compared to
MOCK (FDR < 1%), the effect was classified as "0", mean-
ing that a significant change of the pRB phosphorylation
state was observed. The results demonstrated that knock-
downs of CDK4, CDK6, Cyclin D1, Cyclin E1, ER-α, c-
MYC, ERBB1, ERBB1_2, and IGF-1R indeed resulted in a
significant hypophosphorylation of pRB.
Simulation of loss-of-function of all three receptors
(ERB1_2_3) had resulted in two stable states for pRB: "1"
and "0". Experimentally, we have shown that knockdown
of all three receptors resulted in pRB = 1 suggesting that
combinatorial targeting of ERBB receptors may not be
beneficial to overcome resistance in de novo trastuzumab
resistant cells (Additional file 2, Table 2).
To confirm the effect of knockdowns on G1/S transition at
the DNA level, we measured incorporation of 7-AAD into
the DNA of single cells by flow cytometry 18 hours after
EGF stimulation (Figure 5B). The fractions of cells in G1-
and in S phases were taken to calculate the G1/S ratio for
each knockdown. We regarded gene knockdowns having
similar effects as MOCK or p21 to be positive for G1/S
transition ("value 1"), while G1/S ratios higher than
MOCK or p21 were considered negative ("value 0"). For
the knockdowns of CDK4, Cyclin D1, Cyclin E1, ER-α and
c-MYC, we thus observed no G1/S transition (Table 2).
Table 2 summarizes the simulation results for the final
state of the pRB protein and the respective experimental
data. Indeed, 12 out of 17 knockdown simulations were
consistent with experimentally measured pRB activity lev-
els (compare columns 1 and 2 of Table 2). The correlation
is even slightly better with 7-AAD data, as the results of 13
out of 17 simulations were consistent with those of the p-
pRB data (compare columns 1 and 3 of Table 2). How-
ever, the simulations of ER-α, c-MYC and Cyclin E1
knockdowns gave results that are inconsistent with both
types of the experimental data, pointing out the limits of
our current model, which we will address in the next sec-
tion. Altogether, our results suggest that Cyclin D1, CDK4,
Cyclin E1, ER-α and c-MYC, but neither the combinatorial
targeting of ERBB family receptors nor of key components
of the MAPK (MEK1) and the survival pathways (AKT1)
should be considered as potential targets for further test-
ing in our de novo trastuzumab resistant model cell sys-
tem.
Table 2: Comparison of simulation results with experimental data (p-pRB protein data and 7-AAD DNA data).
Simulated
p-pRB data
7-AAD data
Improved rules
MOCK
1
1
1
1
AKT1
1
1
1
1
MEK1
1
1
1
1
CDK2
1
1
1
1
CDK4
0
0
0
0
p21
1
1
1
1
p27
1
1
1
1
Cyclin D1
0
0
0
0
ERBB2_3
1
1
1
1
ERBB1_3
0/1
1
1
0/1
ERBB1
1
0
1
1
ERBB1_2
0/1
0
1
0/1
IGF1R
1
0
1
1
CDK6
0
0
1
0
ER-α
1
0
0
0
c-MYC
1
0
0
0
Cyclin E1
1
0
0
1
The first column ("Simulated") corresponds to the results of knockdown simulations for the initial logical model, while the last column ("Improved
rules") displays the results obtained with the revised model (improved logical rules for Cyclin D1 as shown in Table 1). In the "Simulated", "p-pRB
data" and "Improved rules" columns, the value "1" denotes the phosphorylation of pRB (G1/S transition) and "0" denotes the lack of
phosphorylation (no G1/S transition). In the "7-AAD data" column, the value "1" denotes a low ratio of G1/S (G1/S transition) and "0" denotes a
high G1/S ratio (no G1/S transition). Bold numbers emphasize the knockdowns for which simulations and both experiments agree. Normal
numbers correspond to the knockdowns for which experiments led to different values, one of them being met by our simulation results. In the case
of ERBB1_3 knockdown, one of the two existing stable states meets the concordant experimental result. The final state depends on the choice of
the initial conditions, but these are difficult to fully specify (a similar situation occurs for the ERBB1_2 knockdown). Finally, Italic numbers denote
the knockdowns for which simulation data differ from the results of both experiments.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 9 of 20
(page number not for citation purposes)
Figure 4 (see legend on next page)
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 10 of 20
(page number not for citation purposes)
Refinement of logical rules and network reconstruction
based on quantitative protein data
To find out whether the observed discrepancies between
our network model and the experimental data were due to
the incorporation of incorrect logical rules, missing inter-
actions, or even missing components in our literature-
based network, we next refined the logical rules and per-
formed a network reconstruction that was based on quan-
titative protein data.
Extracting information about combinatorial regulatory
effects of different proteins affecting a given component is
much more difficult than extracting information about
individual interactions from the literature. We have thus
systematically evaluated modifications of the logical rule
with respect to model prediction capacity. In particular,
the discrepancy between the simulation results and exper-
imental counterparts for c-MYC and ER-α knockdowns
could be solved by changing the logical rules associated
with the Cyclin D1 node. We tested several combinations
for the logical rules on ER-α, c-MYC and Cyclin D1. A
minor modification of the model enabled us to recover
the correct behaviour for the two loss-of-functions: ER-α
and c-MYC (with a stable state having pRB = 0), while
conserving all the behaviour for all other proteins. The
original rules assumed that the presence of one activator
is sufficient (the OR connecting all 4 variables in Table 2).
This is the loosest rule that can be defined for a node that
is activated by several regulators. The modified rule (Cyc-
lin D1 = 1 when ER-α AND c-MYC AND (AKT1 OR MEK1)
is more restrictive as this states that that ER-α AND c-MYC
together with AKT1 OR MEK1, are required to activate
Cyclin D1. The biological implication of this change is
that ER-α, c-MYC and (MEK1 or AKT1) proteins should
act together to make the cells pass through S-phase and
proliferate. In addition, although both transcription fac-
tors are necessary at the same time, the function of the one
of the signaling molecule, AKT1 or MEK1, can be compen-
sated in the cell, but not of the two at the same time. So,
our results may propose a more comprehensive logic for
the regulation of Cyclin D1 in our model cell system.
These results may hint that control of Cyclin D1 is a
sequential event (AKT1 or MEK1 → ER-α → c-MYC) and
can exclude the alternative edges from ER-α, MEK1 or
AKT1 to Cyclin D1. We are thus left with just one knock-
down simulation (Cyclin E1) disagreeing with the experi-
mental data.
In the next step, we wanted to test if the discrepancy
observed in the case of Cyclin E1 knockdown could be
attributed to some missing interactions among the regula-
tory components considered in our logical model. In
order to address this discrepancy and also to examine cell
line specific regulations, we further quantified the activa-
tion and expression levels of most of the network ele-
ments for individual and combinatorial network protein
knockdowns using reverse phase protein arrays. In total,
we quantified the changes in expression of nine network
proteins, as well as the phosphorylation levels of ERK1/2
and AKT1. Some proteins could not be included in these
measurements because of the lack of antibodies suitable
for RPPA. As for the pRB experiments, we examined the
effect of EGF stimulation on the other network proteins
for each knockdown, compared to MOCK. The heatmaps
in Figure 6 show the significant changes, at the expression
or activation level of the proteins before EGF stimulation
(Figure 6A) and 12 h after EGF stimulation (Figure 6B).
Expression and phosphorylation levels either confirmed
known interactions or inferred novel ones (Figure 6B).
The resulting interactions define the network presented in
Figure 7. A jackknife procedure (see Methods section) was
used to eliminate putative indirect edges, which could be
explained by a path along other edges, and only edges
having a jackknife probability greater than 50% were kept
(Figure 7). In the graph, solid black arrows indicate
inferred direct or indirect interactions which are also sup-
ported by published data, whereas the dotted grey arrows
denote novel regulations having been identified for the
HCC1954 reference cellular system. As a result, we
inferred most of the interactions considered in our litera-
ture-based network from the experimental data for trastu-
zumab resistant HCC1954 cells, although some of them
have opposite directions. One should also take into
account that drawing edge directions from biological liter-
Determination of knockdown efficiency and specificity of siRNAs in the HCC1954 cell system
Figure 4 (see previous page)
Determination of knockdown efficiency and specificity of siRNAs in the HCC1954 cell system. A. qRT-PCR
results showing the knockdown efficiency of 20 nM pools of four siRNAs for each gene in the network (50 nM siRNA for
ESR1). ACTB and HPRT1 were used as house-keeping controls. MOCK stands for the samples which were treated only with
Lipofectamine transfection reagent. B. Western blots for the determination of knockdowns for the network elements at pro-
tein level. Since dimers of ERBB family members are accepted as functional units, we used combinatorial RNAi (knockdown of
two genes simultaneously) to produce knockdowns of such dimers. C. qRT-PCR results showing the knockdown efficiency and
effect of one member of ERBB receptor family siRNA on the other two members of the family. Both pools of four individual
siRNAs per gene and only one individual siRNA per gene were used. The concentration of siRNAs was 20 nM. ACTB was used
as house-keeping control. D. Western blots showing the knockdown efficiency and cross reactivity of siRNAs at protein level.
β-actin was taken as loading control.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 11 of 20
(page number not for citation purposes)
ature is usually a daunting task. Indeed, edge directions,
when indicated, are often not well defined or even errone-
ous. Additionally, there can be cell line specific differences
or thus far unknown feedback mechanisms, for example a
feedback from Cyclin D1 to MEK1 or from ER-α to AKT1.
It is, therefore, not really unlikely to see edges that link in
the direction opposite to the expected. Hence, although
this approach demonstrates the feasibility of network
reverse engineering at protein level using robust and
quantitative protein array data, the resulting network was
no help to solve the discrepancy observed for Cyclin E1
knockdown, thereby leaving a gap in our knowledge in
the regulation or regulatory effects of this component. To
sum up, our network inference approach provided us with
the knowledge that the most regulations which were
obtained from the literature were also present in our tras-
tuzumab cell system and novel regulations might have
potential role in the observed phenotype of the cells.
Combinatorial targeting of c-MYC or EGFR in combination
with ERBB2 using small chemical inhibitors in sensitive and
resistant cells
In order to verify the RNAi results and to validate the
potential targets in our de novo resistant cell system, we
applied small chemical inhibitors against c-MYC (10058-
F4) and EGFR (gefitinib), alone and in combination with
trastuzumab, and examined the growth of trastuzumab
resistant HCC1954 cells compared to sensitive SK-BR-3
and BT474 cells. Administration of the c-MYC inhibitor
alone resulted in reduced pRB phosphorylation in all
three cells lines (Figure 8A, left panel), and its application
alone or in combination with trastuzumab also reduced
the growth of these cell lines (Figure 8B, middle panel).
The results were verified using real-time impedance meas-
urements over four days (Figure 8C, right panel), provid-
ing a time-lapse profile of the growth rates. The resulting
data demonstrate that the reduced growth rates of cells
treated with the c-MYC inhibitor was independent from
trastuzumab resistance and thus support the RNAi results
shown in Figure 5A and 5B.
Because combinatorial targeting of ERBB receptors is
already in clinical use (e.g. lapatinib), we next targeted
ERBB1 and ERBB2 receptors in single and combinatorial
settings and compared the outcome in the de novo resist-
ant HCC1954 cell line with the trastuzumab sensitive cell
lines. First, we examined the downstream effectors of
ERBB receptors (ERK1/2 and AKT1) and of pRB after treat-
ment with trastuzumab or gefitinib (targets ERBB1) (Fig-
ure 8B, left panel). In HCC1954 cells, no reduction in the
expression levels of EGFR/ERBB2 or phosphorylation lev-
els of their downstream signal mediators was observed for
both treatments. However, reduced pRB phosphorylation
was observed in gefitinib-treated HCC1954 cells. Both,
the WST-1 viability assay and real-time impedance meas-
urements demonstrated that trastuzumab resistant
HCC1954 cells were also resistant to gefitinib treatment
alone or in combination with trastuzumab. (Figure 8B,
middle and right panel).
In SK-BR-3 cells, AKT1 and pRB phosphorylation levels
were lower after trastuzumab treatment, and pRB phos-
phorylation was reduced after incubation with gefitinib.
In BT474 cells, a strong reduction in AKT and ERK1/2
phosphorylation was measured after gefitinib treatment,
while no such an effect was evident for trastuzumab treat-
ment (Figure 8B, left panel). Real-time impedance meas-
Analysis of the effects of knockdowns on G1/S transition in
trastuzumab resistant HCC1954 cells
Figure 5
Analysis of the effects of knockdowns on G1/S transi-
tion in trastuzumab resistant HCC1954 cells. A. Phos-
phorylation state of pRB (output of the network) after 12
hour EGF stimulation (input of the network). Box plots show
the quantitative phosphorylation of pRB protein for the
knockdown of each network protein compared to MOCK
(only transfection reagent, no siRNA). B. G1/S ratio after 18
hours of EGF stimulation for corresponding knockdowns
compared to MOCK (solid line) and p21 knockdown (dotted
line) using 7-AAD DNA staining.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 12 of 20
(page number not for citation purposes)
urement showed for the SK-BR-3 cells that combinatorial
targeting of EGFR and ERBB2 had a strong additive effect
to reduce cell proliferation (Figure 8B, right panel). This
additive effect was also visible for BT474 cells although it
was not as strong as in SK-BR-3 cells. These data support
our RNAi results, suggesting that the combinatorial target-
ing of the EGFR and ERBB2 with gefitinib and trastuzu-
mab, respectively, might not be effective to sensitize the
cells to trastuzumab treatment in de novo trastuzumab
resistance. However, these drugs in combination might
lead to an improved outcome in sensitive cells, and poten-
tially also in tumors, as compared to applying them indi-
vidually.
Discussion
In the present study, we have applied a systems biology
approach to identify alternative targets in de novo trastuzu-
mab resistant breast cancer. While several studies have
dealt with mechanisms leading to acquired trastuzumab
resistance, there has been no comprehensive study that
searched for targets alternative to ERBB2 in de novo trastu-
zumab resistance. Since the aim of cancer therapy is to
reduce the growth rate of cancer cells, and trastuzumab
resistant breast cancer cells escape cell cycle arrest during
treatment, we focused on a protein network that connects
ERBB signaling to G1/S phase transition, in order to deter-
mine new potential targets for perturbation. In contrast to
previous studies, which had focused on the involvement
of an individual protein in the resistant phenotype of the
cells, we aimed to examine the roles of each protein in the
context of their interactions at a protein network level.
Several modeling studies about the ERBB receptor-regu-
lated signaling pathways have been published recently
[30,31]. These studies considered the activation of key
intermediates (ERK1/2 and AKT) upon EGF and HRG
stimulation and proposed differential dynamical models
for these pathways. Likewise, various models have been
proposed for the control of the mammalian cell cycle
[18,22,32]. In our study, we combined these two cellular
processes into one coherent network to find novel strate-
gies for breast cancer therapy. First, we derived a logical
network from published data (Figure 3 and Table 1). Sys-
tematic simulations of loss-of-function perturbations
were performed, and the final state of pRB phosphoryla-
tion (the marker for G1/S transition) was determined in
each case. These computational results were then com-
pared with experimental knockdowns, obtained by RNA
Response of network proteins for corresponding knockdowns in trastuzumab resistant HCC1954 breast carcinoma cells (A)
before and (B) after EGF stimulation
Figure 6
Response of network proteins for corresponding knockdowns in trastuzumab resistant HCC1954 breast carci-
noma cells (A) before and (B) after EGF stimulation. Heatmaps were drawn for a false discovery rate (FDR) of less
than 1%.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 13 of 20
(page number not for citation purposes)
interference. While each network component was targeted
by siRNAs in a single knockdown setting, both constitu-
ents of ERBB heterodimers were repressed in a combina-
torial RNAi setting (Figure 4). We quantified the effects of
these knockdowns on pRB phosphorylation with reverse
phase protein arrays (RPPA) (Figure 2 and Figure 5).
Knockdowns of c-MYC, ER-α and Cyclin E1 resulted in a
very strong reduction in pRB phosphorylation compared
to ERBB1, ERBB1/ERBB2, and IGF1R knockdowns (Figure
5A). We then determined the ratio of G1/S to further ver-
ify the effect of these knockdowns on G1/S transition (Fig-
ure 5B). DNA staining enabled us to differentiate the
strong effects of c-MYC, ER-α and Cyclin E1 knockdown
as compared to weaker effects of ERBB1, ERBB1/ERBB2,
and IGF1R knockdowns (Figure 5B).
Consequently, Cyclin D1 and CDK4 were identified as
potential targets from both simulation and experiments.
This result was expected, as in response to an external
stimulus, Cyclin D1 and CDK4 make a complex that
Tentative regulatory interactions inferred from protein expression and activation data for trastuzumab resistant HCC1954
cells
Figure 7
Tentative regulatory interactions inferred from protein expression and activation data for trastuzumab resist-
ant HCC1954 cells. The heatmaps in Figure 6B were used to infer the interactions of proteins in the HCC1954 cell system
with a probability higher than 50%. The numbers next to each arrow indicate the probability of each interaction. Solid black
arrows denote the interactions (both direct and indirect) supported by published data, whereas dotted grey arrows denote
novel interactions (compare with Figure 3). Normal arrows denote positive regulations, whereas blunt arrows denote negative
regulations.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 14 of 20
(page number not for citation purposes)
Figure 8 (see legend on next page)
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 15 of 20
(page number not for citation purposes)
phosphorylates pRB, and which, in turn, enables G1/S
transition. Accordingly, we found knockdown of these
proteins to result in a significant reduction in the phos-
phorylation of pRB. In contrast, c-MYC, ER-α and Cyclin
E1 were identified by experimental analyses on de novo
trastuzumab resistant cells, but had not been predicted in
the initial network model. After network refinement, c-
MYC and ER-α were also predicted as targets from the
model (Table 1 and 2). Hence, we demonstrated that this
approach enables the reconstruction of phenotype-spe-
cific interactions, which are essential to predict therapeu-
tic strategies.
In addition, missing components in the protein network
can also be inferred. For example, while Cyclin E1 and
CDK2 form a complex, which further phosphorylates the
pRB protein, our experimental data show that only loss of
Cyclin E1, but not of CDK2, significantly repressed phos-
phorylation of pRB (Figure 5A). This result suggests that
CDK2 could be a dispensable component for the G1/S
transition in de novo trastuzumab resistant breast cancer,
as it has previously been shown for colon cancer cells [33].
This observation raises the question which alternative
interaction partners of Cyclin E1 could promote G1/S
transition.
In our study, the transcription factors c-MYC and ER-α
were identified as potential targets to overcome de novo
trastuzumab resistance. Park et al had previously shown
that an amplification of the c-MYC gene is correlated with
ERBB2 overexpression in breast cancer [34]. In trastuzu-
mab sensitive cells, ERBB2-targeted antibodies can inhibit
c-MYC through inhibition of the MAPK and AKT pathway
which, in turn, increases the activity of p27 towards the
CDK2-Cyclin E complex [35]. Here, we demonstrated that
loss of c-MYC activity results in a reduction of the CDK4
level which then results in reduced pRB phosphorylation
(Figure 7). Targeting c-MYC with a specific chemical
inhibitor alone or in combination with trastuzumab also
resulted in a strong reduction in pRB phosphorylation
and cell growth, both in trastuzumab resistant and sensi-
tive cells (Figure 8A). We conclude that targeting c-MYC
alone or in combination with trastuzumab could be an
interesting candidate for a clinical trial. Cross-talk
between ERBB2 signaling and ER-α activation has been
previously reported [36], and an increase in the ERBB2
expression level has been reported in tamoxifen resistant
cells [37]. In this study, we have shown that ER-α is
another possible target in ERBB2 overexpressing and tras-
tuzumab resistant HCC1954 cells. This suggests an inter-
play between ER-α and ERBB2 receptors in the context of
bypassing the effects of drug treatment.
Interestingly, the five novel candidates to be targeted in de
novo trastuzumab resistant breast cancer have one feature
in common: they all either directly (ER-α, c-MYC and
CDK4) or indirectly (Cyclin D1 and Cyclin E1) regulate
the p27 protein, which plays a key role also in acquired
trastuzumab resistance [38]. In addition, our study indi-
cated that combinatorial targeting of either of ERBB1,
ERBB2 and ERRB3 may not enhance sensitivity to trastu-
zumab in de novo resistant patients, although ERBB pro-
teins have been previously considered as promising
targets. These results let us hypothesize that these cell sur-
face proteins (here: ERBB receptors or IGF1R) are decou-
pled from intracellular processes (here: G1/S transition)
in the de novo trastuzumab resistant cell system. Targeting
EGFR alone or in combination with ERBB2 further sup-
ported this notion. While trastuzumab sensitive cells (SK-
BR-3 and BT474) were responding to gefitinib as well as
combination of gefitinib and trastuzumab treatment in an
additive manner, de novo trastuzumab resistant cells
(HCC1954) did not respond at all (Figure 8B). This obser-
vation suggests that combinatorial targeting of cell surface
receptors might be beneficial as it is an uncommon per-
turbation for cells [39], but it should be taken into consid-
eration that this might be cell system- or patient specific.
Effects of c-MYC inhibitor 10058-F4 and EGFR inhibitor gefitinib alone or in combination with trastuzumab on the viability of
resistant (HCC1954) and sensitive (SK-BR-3 and BT474) cells
Figure 8 (see previous page)
Effects of c-MYC inhibitor 10058-F4 and EGFR inhibitor gefitinib alone or in combination with trastuzumab on
the viability of resistant (HCC1954) and sensitive (SK-BR-3 and BT474) cells. A. Left panel: Western blot data show-
ing the phosphorylation state of pRB protein after treatment with c-MYC inhibitor (10058-F4, 80 μM) for 24 hours. Middle
panel: WST-1 viability assay over 4 days to determine the response of cells to c-MYC inhibitor alone and in combination with
trastuzumab. DMSO is used as vehicle control. Right panel: Impedance measurement for real-time determination of cell growth
before (24 hours) and after (72 hours) treatment of cells with c-MYC inhibitor, trastuzumab, and the combination of both
drugs. The impedance measurements were normalized to the time point where we have added the drugs. The vertical line
demonstrates the time point of normalization. B. Left panel: Western blot data showing the effect of different concentrations of
trastuzumab and gefitinib (1 μM) on the expression levels of EGFR and ERBB2 and on the phosphorylation states of ERK1/2,
AKT and pRB proteins. H2O is vehicle control for trastuzumab and DMSO is for gefitinib. Middle panel: WST-1 assay to deter-
mine the response of cells to gefitinib and gefitinib + trastuzumab. Right panel: The impedance measurement for real-time meas-
urement of cell growth before (24 hours) and after (72 hours) treatment of cells with gefitinib, trastuzumab and combination of
both drugs. Normalization of cell index is as in A.
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 16 of 20
(page number not for citation purposes)
According to our results, targeting EGFR with siRNAs
alone resulted in an efficient knockdown at the mRNA
level; however, no reduction was observed at the protein
level (Figure 4). This phenomenon might be explained by
the stabilization of EGFR after dimerization with the over-
expressed ERBB2 receptor. Complementary to this obser-
vation, knocking down ERBB2 resulted in reduced EGFR
expression at the protein level, although no reduction of
EGFR was observed at the mRNA level (Figure 4). These
data demonstrate an explicit dependence of EGFR protein
abundance on ERBB2 expression, and should be kept in
mind when EGFR is targeted in cancer therapies. This
observation is independent of trastuzumab sensitivity,
but is highly influenced by the ERBB2 expression level
(Figure 1 and Additional file 1, Figure 3).
It should be noted that the logical formalism used in this
work clearly caricatures subtle dose effects into all-or-
none responses for all the components considered. The
resulting logical model should thus be taken as a first step
in the formalisation of the regulatory network involved in
trastuzumab resistance. However, it is appropriate to
translate qualitative information and compare the behav-
iour of alternative network wirings or logical rules with
data sets for unperturbed and perturbed situations. Once
the regulatory wiring and the logical rules be reasonably
established, it will be possible to take advantage of multi-
level logical modeling extensions, or yet to translate our
Boolean models into more quantitative formalisms (e.g.
ordinary differential equations, or yet hybrid or stochastic
Petri nets).
Conclusion
We constructed a literature-based protein network and
combined computational simulations, validation experi-
ments using RNAi, as well as chemical inhibitors, and net-
work inference based on proteomic data, in order to
identify novel targets with potential for individual and
combinatorial therapies in breast cancer. Our concept to
combine experimental and computational biology dem-
onstrated the strengths and limitations of using literature-
based models for simulations of therapeutic strategies.
Furthermore, this study led us to select c-MYC as a candi-
date to be tested in in vitro and in vivo models, regarding
future treatments for breast cancer which is de novo resist-
ant to trastuzumab. Our results also suggest that combina-
torial targeting of key ERBB receptors might have better
outcome than individual therapies in trastuzumab sensi-
tive cells, but not in de novo trastuzumab resistant cells.
Methods
Cell Culture
Five human breast cancer cell lines (HCC1954, SK-BR-3,
MDA-MB-231, BT474 and MCF-7) as well as the normal
breast epithelial cell line MCF-12A were obtained from
ATCC (Manassas, VA). HCC1954 cells (CRL-2338) were
cultured in RPMI 1640 Modified Medium (ATCC), SK-BR-
3 cells (HTB-30) in McCoy's 5a medium (GIBCO BRL),
and MDA-MB-231 cells (HTB-26) in Leibovitz's L-15
medium (Sigma). BT474 cells (HTB-20) were cultured in
DMEM medium, MCF-7 cells (HTB-22) in Eagle's Minim-
ial essential medium, and MCF-12A cells (CRL-10782) in
a medium containing a 1:2 mixture of Dulbecco's Modi-
fied Eagle's Medium and Ham's F12 medium. All media
were supplemented with 50 U/mL penicillin, 50 μg/mL
streptomycin sulphate, 1% non-essential amino acids and
10% fetal bovine serum (all media and supplements from
Gibco BRL). Additionally, 2.2 g/L sodium bicarbonate
was supplemented for MDA-MB-231 cells. Media for
BT474 cells were supplemented with 10% NCTC
medium, 500 μl bovine insulin and 100 μl Oxalic acid, for
MCF-7 cells we added 0.01 mg/mL bovine insulin, and
such for MCF-12A cells were supplemented with 20 ng/
mL EGF, 100 ng/mL cholera toxin, 0.01 mg/mL bovine
insulin, and 500 ng/mL hydrocortisone. The cells were
incubated at 37°C with 5% CO2 and split 2–3 times per
week in a 1:3 ratio for no more than 20 passages. All cell
lines were validated by genotyping.
3-D cell culture
HCC1954 and SK-BR-3 cells were cultured in 8-well
chamber slides in order to examine the effect of trastuzu-
mab on proliferation. Geltrex (Invitrogen, Carlsbad, CA)
was thawed on ice and 40 μL was pipetted per well and left
for 30 min at 37°C to solidify. HCC1954 and SK-BR-3
(5.000 cells/well) cells were seeded in medium supple-
mented with 1:50 Geltrex and EGF (BD Biosciences), and
with or without trastuzumab (100 nM) (Roche, Penzberg,
Germany). The cells were incubated at 37°C and 5% CO2
for eight days, and medium was changed after 4 days.
siRNA transfections and EGF stimulations
HCC1954 cells were seeded at a number of 7 × 105 cells
per 10 cm petri dish in antibiotic free medium. Conflu-
ency of the cells was 50–60% at the day of transfection.
Sequences of siRNAs are given in Additional file 1, Table
2. Twenty nM of siRNA (except ESR1 siRNA (50 nM))
(Dharmacon, Lafayette, CO) and 25 μL of Lipofectamine
2000™ (Invitrogen, Carlsbad, CA) were diluted separately
in reduced-serum medium OptiMEM (Gibco BRL) and
incubated for 5 minutes at RT. The two solutions were
then mixed and incubated for 20 minutes at RT. The
siRNA-Lipofectamine 2000™ mixture was then added to
the cells and the dishes were shaken by gentle rocking.
MOCK transfected cells were treated with Lipofectamine
2000, but no siRNA was added. The cells were incubated
at 37°C and 5% CO2 for 24 hours. After incubation, cells
were starved by Dif-3 (30 μM) (Sigma) for 22 hours in
medium containing 10% FBS. Cells were further starved
in 0% FBS medium for 2 hours. After 24 hours of starva-
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 17 of 20
(page number not for citation purposes)
tion, cells were stimulated with EGF (25 ng/mL) for 6, 12,
18 and 24 hours.
Cell lysis and Western blotting
At each time point, medium was removed and cells were
washed with ice-cold PBS containing 10 mM NaF and 1
mM Na4VO3. Lysis of cells was performed on ice by scrap-
ing or by cold trypsinization, and shaking on over-head
shaker for 15 minutes at 4°C with 70 μl M-PER lysis buffer
(Pierce, Rockford, IL) containing protease inhibitor Com-
plete Mini (Roche, Basel), anti-phosphatase PhosSTOP
(Roche, Basel), 10 mM NaF and 1 mM Na4VO3. Protein
concentrations were determined with a BCA Protein Assay
Reagent Kit (Pierce, Rockford, IL). Proteins were denatur-
ated with 4× Roti Load (Roth, Karlsruhe, Germany) at
95°C for 5 minutes, and 12 μg proteins were loaded in
every lane. Protein samples were separated by 8% or 12%
SDS PAGE, electroblotted to PVDF membranes (Amer-
sham Biosciences, USA) and exposed to primary antibod-
ies. A list of antibodies is given in Additional file 1, Table
3, together with their dilutions. Horseradish peroxidase
conjugated anti-mouse or rabbit antibodies (Amersham
Biosciences, USA) were used as secondary antibodies and
signals were detected by enhanced chemiluminescence
(Amersham Biosciences, USA).
TaqMan (qRT-PCR)
Total RNA was extracted from the cells by using the Invi-
sorb Spin cell RNA mini kit (Invitek GmbH, Berlin, Ger-
many), and single-stranded cDNA was transcribed with
the RevertAid H Minus First Strand cDNA Synthesis kit
(Fermentas, St. Leon-Rot, Germany). Ten nanograms of
total RNA were used for each reaction. qRT-PCR for target
genes and housekeeping genes ACTB and HPRT1 was per-
formed with the ABI Prism 7900HT Sequence Detection
System (Applied Biosystems, Weiterstadt, Germany),
applying probes of the Universal Probe Library (Roche,
Penzberg, Germany). Primers were synthesized by MWG
(Ebersberg, Germany). Sequences of primers and the
respective UPL probe numbers are given in Additional file
1, Table 4.
Reverse Phase Protein Arrays (RPPA)
Cell lysates were prepared as for Western Blotting. All
lysates were adjusted to a total protein concentration of 3
μg/μl. Cell lysates were mixed 1:2 with 2× Protein Array-
ing Buffer (Whatman, Brentfort, UK) to yield a final pro-
tein concentration of 1.5 μg/μL. The samples were printed
with a non-contact piezo spotter, sciFlexxarrayer S5 (Sci-
enion, Berlin, Germany), in four replicate spots per sam-
ple and subarray, and two subarrays per slide onto
nitrocellulose coated ONCYTE-slides (Grace Bio Labs,
Bend, USA). Twenty replicate slides were produced per
run. Approximately 2.25 ng total proteins were delivered
per spot. As spotting control, the total protein content of
all spots was determined for two replicate slides with the
FAST Green FCF assay. All antibody signals were normal-
ized according to their total protein content. Slides were
blocked over night and target proteins were detected with
specific primary antibodies (Additional file 1, Table 3)
using a protein array incubation chamber (n1-quadrat,
Metecon, Mannheim, Germany). Detection of primary
antibodies was carried out with near-infrared (NIR)-dye
labeled secondary antibodies and visualized using an
Odyssey scanner (LI-COR, Lincoln, USA). Signal intensi-
ties were quantified using Odyssey 2.0 software, corrected
for spot-specific background signals and normalized for
their total protein concentrations.
7-AAD staining and analysis
Directly after siRNA transfections, cells were synchronized
for 24 hours (see above), and stimulated with EGF for 18
hours. Then, cells were trypsinized, washed once with PBS
and centrifuged. Ice cold methanol was added to the cell
pellets while vortexing the FACS tube. After incubation of
cells at -20°C overnight, methanol was removed and 250
μl of 7-AAD (1:40 dilution) (Calbiochem, Darmstadt,
Germany) was added to each tube and incubated for 1.5
hours at 4°C in the dark. The measurement was done by
flow cytometry (FACS Calibur, BD Biosciences) using the
FL3 channel for 7-AAD staining. Analysis of the 7-AAD
results was performed using CellQuest Pro software with
the histogram statistics option and a gate on the main cell
population.
WST-1 cell viability assay
HCC1954 cells were seeded at a number of 2,500 cells/
well in 96 well format in 100 μL of 10% FBS medium
without antibiotics. After 24 hours of incubation, cells
were washed once with PBS and then incubated with 200
μl of either trastuzumab (100 nM) (Roche, Penzberg, Ger-
many) or gefitinib (1 μM) (Biaffin, Kassel, Germany) or c-
MYC inhibitor 10058-F4 (80 μM) (Sigma) containing
10% FBS medium. Cells were incubated for 24 hours and
each day 20 μl of WST-1 reagent (Roche, Basel) was pipet-
ted to the cells. Absorbance was measured at 450 nm after
2.5 hours with a SpectraMAX 190 (Molecular Devices,
UK). The WST-1 assay was performed over four days with
one measurement taken on everyday.
Real-time cell-electrode impedance measurements
One hundred microliters of growth medium was added to
the wells of E-plates (Roche, Penzberg) for background
measurements. Then, 100 μL of HCC1954, SK-BR-3 and
BT474 cell suspensions were added at a number of 8,000
(for HCC1954 cells) and 10000 (for SK-BR-3 and BT474)
cells/well. E-plates were incubated at room temperature
for 30 minutes; then transferred to the holder and incu-
bated at 37°C with 5% CO2. The continuous impedance
measurement was recorded and converted to a cell index
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 18 of 20
(page number not for citation purposes)
(CI). After 24 hours, the chemical inhibitors (10058-F4
(80 μM) and gefitinib (1 μM) or/and trastuzumab (100
nM) were added to the respective wells and impedance
measurements were continued for 72 hours. Results were
analyzed using RTCA Software 1.0 (Roche, Penzberg, Ger-
many).
Modeling, simulations and data analysis
Logical modeling and simulations
A logical model is defined by a regulatory graph, where the
nodes and arcs represent the regulatory components and
interactions, respectively. The dynamical behavior of each
component is then defined by logical functions (also rep-
resented in terms of logical parameters), which associate a
target value for this component depending on the level of
its regulators. The dynamics of the system is represented
in terms of a state transition graph, where the nodes
denote states of the system (i.e., a vector giving the levels
of activity of all components), and the arcs denote state
transitions (i.e., a change in the value of one or several
component(s), depending on the values of the relevant
logical functions or parameters). In state transition
graphs, terminal nodes correspond to "stable states". Note
that, for most of the conditions considered, our ERBB
receptor regulated G1/S model has a single stable state.
The Boolean model of ERBB signaling network was
defined and analyzed using the GINsim software [22,23].
Beginning with relevant initial states, simulations using
the logical rules defined in Table 1 was performed. For
MOCK case, the initial levels of EGF, all ERBBs, p21 and
p27 were set to 1, whereas Cyclin D1 was set to 0. The ini-
tial levels of the other proteins were left undefined, mean-
ing that both possible levels were considered. A
knockdown can be simulated in GINsim by setting the
corresponding protein's initial level and its maximal value
to 0. The resulting parameterized model and all simula-
tions can be downloaded from the model repository
referred at the GINsim web page: http://gin.univ-mrs.fr/
GINsim/model_repository.html.
Analysis of knockdown responses
Statistical significance of protein expression changes and
pRB phosphorylation due to knockdowns via RNA inter-
ference were calculated using the ANOVA method: protein
expression ~ knockdown effect + biological replicate factor +
error A multiple testing correction was performed using
Benjamini-Hochberg's method [40] with a false discovery
rate (FDR) significance cut off of 1%.
Network inference
Whenever a knockdown significantly affected the expres-
sion of another protein with an FDR < 1%, an edge was
drawn. Then a transitive reduction of the graph was calcu-
lated (i.e. eliminating putative indirect edges, which could
also be explained by another path in the graph [41]. Since
the transitive reduction for graphs with cycles is not
unique and depends on the ordering of the nodes, we
implemented a jackknife procedure, i.e. we left out each
node once, estimated the network, and finally counted for
each edge the frequency of the occurrence among all jack-
knife samples. The corresponding jackknife probability is
reported at each edge. We also performed the multiple
testing corrections separately within each sample of the
jackknife procedure, since the false discovery rate depends
on the distribution of all raw p-values, which may change
with the differing gene selection in each jackknife sample.
The R source code is available from the authors upon
request. Only the edges having a jackknife probability
greater than 50% were kept.
Abbreviations
Dif-3: Dictyostelium differentiation-inducing factor-3;
EGF: Epidermal Growth Factor; FDR: False discovery rate;
qRT-PCR: Quantitative real-time polymerase chain reac-
tion; RNAi: RNA interference; siRNA: small interfering
RNA; 3-D cell culture: Three dimensional cell culture; 7-
AAD: 7-Aminoactinomycin
Authors' contributions
ÖS, TB and DA designed the research; ÖS, CL, JM and SB
performed the research; ÖS, TB, HF, CC and DT carried
out computational modeling and simulations; IS and SW
participated in sequence analysis; ÖS, HF, CL, UK, MM,
CC, DT, TB and DA analyzed data; ÖS, HF, CC, DT, AP,
SW, TB and DA wrote the manuscript. All authors read
and approved the final manuscript.
Additional material
Additional file 1
This folder contains the following items: 1. Figure 1 and its figure leg-
end (Page 1). 2. Figure 2 and its figure legend (Page 2). 3. Figure 3 and
its figure legend (Page 3). 4. Table 1: List of references for Figure 3 (Page
4) 5. Table 2: List of siRNAs and their sequences (Page 5) 6. Table 3:
List of antibodies (Page 6). 7. Table 4: List of primers, their sequences and
probe numbers (Page 7).
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-3-1-S1.pdf]
Additional file 2
This folder contains the following items: 1. Table 1: Stable states and
pRB response for single and multiple knockdowns of network proteins
(Pages 1–13). 2. Table 2: Analysis of the effects of knockdowns on G1/S
transition (p-pRB response) (Page 13).
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-3-1-S2.pdf]
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 19 of 20
(page number not for citation purposes)
Acknowledgements
The authors would like to thank Ute Ernst and Christian Schmidt for excel-
lent technical assistance, as well as Dirk Ledwinka for IT support. Special
thanks to Roche (Penzberg, Germany) for providing us with trastuzumab
and early access to the Xcelligence screening system.
This project was supported by the German Federal Ministry of Education
and Research (BMBF) within National Genome Research Network Pro-
gram Grants IG-Cellular Systems Genomics, and IG Prostate-Cancer. Fur-
ther support was from the Helmholtz Program SB-Cancer as well as the EU
FP6 TRANSFOG project (contract LSHC-CT-2004-503438). DT and CC
acknowledge the support of the French Ministry of Research (ANR project
JC05-53969), of the EU FP6 DIAMONDS STREP (contract LSHG-CT-
2004-503568), and of the Belgian IAP BioMaGNet project for the develop-
ment of the GINsim software.
This article is dedicated to the memory of Professor Annemarie Poustka,
who was the founder and head of the Division Molecular Genome Analysis
at the DKFZ. She was an inspiring scientist and a wonderful person.
References
1.
Singh Y, Palombo M, Sinko PJ: Recent trends in targeted antican-
cer prodrug and conjugate design. Curr Med Chem 2008,
15(18):1802-1826.
2.
Piccart M: Circumventing de novo and acquired resistance to
trastuzumab: new hope for the care of ErbB2-positive breast
cancer. Clin Breast Cancer 2008, 8(Suppl 3):S100-113.
3.
Marmor MD, Skaria KB, Yarden Y: Signal transduction and onco-
genesis by ErbB/HER receptors. Int J Radiat Oncol Biol Phys 2004,
58(3):903-913.
4.
Holbro T, Civenni G, Hynes NE: The ErbB receptors and their
role in cancer progression. Exp Cell Res 2003, 284(1):99-110.
5.
Klapper LN, Glathe S, Vaisman N, Hynes NE, Andrews GC, Sela M,
Yarden Y: The ErbB-2/HER2 oncoprotein of human carcino-
mas may function solely as a shared coreceptor for multiple
stroma-derived growth factors. Proc Natl Acad Sci USA 1999,
96(9):4995-5000.
6.
Citri A, Yarden Y: EGF-ERBB signalling: towards the systems
level. Nat Rev Mol Cell Biol 2006, 7(7):505-516.
7.
Zaczek A, Brandt B, Bielawski KP: The diverse signaling network
of EGFR, HER2, HER3 and HER4 tyrosine kinase receptors
and the consequences for therapeutic approaches. Histol His-
topathol 2005, 20(3):1005-1015.
8.
Vogel CL, Cobleigh MA, Tripathy D, Gutheil JC, Harris LN, Fehren-
bacher L, Slamon DJ, Murphy M, Novotny WF, Burchmore M, Shak S,
Stewart SJ, Press M: Efficacy and safety of trastuzumab as a sin-
gle agent in first-line treatment of HER2-overexpressing
metastatic breast cancer. J Clin Oncol 2002, 20(3):719-726.
9.
Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A,
Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L:
Use of chemotherapy plus a monoclonal antibody against
HER2 for metastatic breast cancer that overexpresses
HER2. N Engl J Med 2001, 344(11):783-792.
10.
Nahta R, Esteva FJ: Herceptin: mechanisms of action and resist-
ance. Cancer Lett 2006, 232(2):123-138.
11.
Cobleigh MA, Vogel CL, Tripathy D, Robert NJ, Scholl S, Fehren-
bacher L, Wolter JM, Paton V, Shak S, Lieberman G, Slamon DJ: Mul-
tinational study of the efficacy and safety of humanized anti-
HER2 monoclonal antibody in women who have HER2-over-
expressing metastatic breast cancer that has progressed
after chemotherapy for metastatic disease. J Clin Oncol 1999,
17(9):2639-2648.
12.
Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE Jr, Davidson NE,
Tan-Chiu E, Martino S, Paik S, Kaufman PA, Swain SM, Pisanski TM,
Fehrenbacher L, Kutteh LA, Vogel VG, Visscher DW, Yothers G,
Jenkins RB, Brown AM, Dakhil SR, Mamounas EP, Lingle WL, Klein
PM, Ingle JN, Wolmark N: Trastuzumab plus adjuvant chemo-
therapy for operable HER2-positive breast cancer. N Engl J
Med 2005, 353(16):1673-1684.
13.
Massague J: G1 cell-cycle control and cancer. Nature 2004,
432(7015):298-306.
14.
Lane HA, Beuvink I, Motoyama AB, Daly JM, Neve RM, Hynes NE:
ErbB2 potentiates breast tumor proliferation through mod-
ulation of p27(Kip1)-Cdk2 complex formation: receptor
overexpression does not determine growth dependency. Mol
Cell Biol 2000, 20(9):3210-3223.
15.
Seville LL, Shah N, Westwell AD, Chan WC: Modulation of pRB/
E2F functions in the regulation of cell cycle and in cancer.
Curr Cancer Drug Targets 2005, 5(3):159-170.
16.
Harbour JW, Luo RX, Dei Santi A, Postigo AA, Dean DC: Cdk phos-
phorylation triggers sequential intramolecular interactions
that progressively block Rb functions as cells move through
G1. Cell 1999, 98(6):859-869.
17.
Grillo M, Bott MJ, Khandke N, McGinnis JP, Miranda M, Meyyappan M,
Rosfjord EC, Rabindran SK: Validation of cyclin D1/CDK4 as an
anticancer drug target in MCF-7 breast cancer cells: Effect of
regulated overexpression of cyclin D1 and siRNA-mediated
inhibition of endogenous cyclin D1 and CDK4 expression.
Breast Cancer Res Treat 2006, 95(2):185-194.
18.
Haberichter T, Madge B, Christopher RA, Yoshioka N, Dhiman A,
Miller R, Gendelman R, Aksenov SV, Khalil IG, Dowdy SF: A systems
biology dynamical model of mammalian G1 cell cycle pro-
gression. Mol Syst Biol 2007, 3:84.
19.
Dyson N: The regulation of E2F by pRB-family proteins. Genes
Dev 1998, 12(15):2245-2262.
20.
Ptitsyn AA, Weil MM, Thamm DH: Systems biology approach to
identification of biomarkers for metastatic progression in
cancer. BMC Bioinformatics 2008, 9(Suppl 9):S8.
21.
Yildirim MA, Goh KI, Cusick ME, Barabasi AL, Vidal M: Drug-target
network. Nat Biotechnol 2007, 25(10):1119-1126.
22.
Faure A, Naldi A, Chaouiya C, Thieffry D: Dynamical analysis of a
generic Boolean model for the control of the mammalian
cell cycle. Bioinformatics 2006, 22(14):e124-131.
23.
Gonzalez AG, Naldi A, Sanchez L, Thieffry D, Chaouiya C: GINsim:
a software suite for the qualitative modelling, simulation and
analysis of regulatory networks. Biosystems 2006, 84(2):91-100.
24.
Sahin O, Lobke C, Korf U, Appelhans H, Sultmann H, Poustka A, Wie-
mann S, Arlt D: Combinatorial RNAi for quantitative protein
network
analysis.
Proc
Natl
Acad
Sci
USA
2007,
104(16):6579-6584.
25.
Loebke C, Sueltmann H, Schmidt C, Henjes F, Wiemann S, Poustka A,
Korf U: Infrared-based protein detection arrays for quantita-
tive proteomics. Proteomics 2007, 7(4):558-564.
26.
Takahashi-Yanaga F, Taba Y, Miwa Y, Kubohara Y, Watanabe Y,
Hirata M, Morimoto S, Sasaguri T: Dictyostelium differentiation-
inducing factor-3 activates glycogen synthase kinase-3beta
and degrades cyclin D1 in mammalian cells. J Biol Chem 2003,
278(11):9663-9670.
27.
Sebastian S, Settleman J, Reshkin SJ, Azzariti A, Bellizzi A, Paradiso A:
The complexity of targeting EGFR signalling in cancer: from
expression
to
turnover.
Biochim
Biophys
Acta
2006,
1766(1):120-139.
28.
Guy PM, Platko JV, Cantley LC, Cerione RA, Carraway KL 3rd:
Insect cell-expressed p180erbB3 possesses an impaired tyro-
sine kinase activity. Proc Natl Acad Sci USA 1994,
91(17):8132-8136.
29.
Pagano M, Tam SW, Theodoras AM, Beer-Romero P, Del Sal G, Chau
V, Yew PR, Draetta GF, Rolfe M: Role of the ubiquitin-proteas-
ome pathway in regulating abundance of the cyclin-depend-
ent kinase inhibitor p27. Science 1995, 269(5224):682-685.
30.
Kumar N, Wolf-Yadlin A, White FM, Lauffenburger DA: Modeling
HER2 effects on cell behavior from mass spectrometry phos-
photyrosine data. PLoS Comput Biol 2007, 3(1):e4.
31.
Birtwistle MR, Hatakeyama M, Yumoto N, Ogunnaike BA, Hoek JB,
Kholodenko BN: Ligand-dependent responses of the ErbB sig-
naling network: experimental and modeling analyses. Mol
Syst Biol 2007, 3:144.
32.
Novak B, Tyson JJ: A model for restriction point control of the
mammalian cell cycle. J Theor Biol 2004, 230(4):563-579.
33.
Tetsu O, McCormick F: Proliferation of cancer cells despite
CDK2 inhibition. Cancer Cell 2003, 3(3):233-245.
34.
Park K, Kwak K, Kim J, Lim S, Han S: c-myc amplification is asso-
ciated with HER2 amplification and closely linked with cell
proliferation in tissue microarray of nonselected breast can-
cers. Hum Pathol 2005, 36(6):634-639.
Publish with BioMed Central and every
scientist can read your work free of charge
"BioMed Central will be the most significant development for
disseminating the results of biomedical research in our lifetime."
Sir Paul Nurse, Cancer Research UK
Your research papers will be:
available free of charge to the entire biomedical community
peer reviewed and published immediately upon acceptance
cited in PubMed and archived on PubMed Central
yours — you keep the copyright
Submit your manuscript here:
http://www.biomedcentral.com/info/publishing_adv.asp
BioMedcentral
BMC Systems Biology 2009, 3:1
http://www.biomedcentral.com/1752-0509/3/1
Page 20 of 20
(page number not for citation purposes)
35.
Le XF, Pruefer F, Bast RC Jr: HER2-targeting antibodies modu-
late the cyclin-dependent kinase inhibitor p27Kip1 via multi-
ple signaling pathways. Cell Cycle 2005, 4(1):87-95.
36.
Gee JM, Robertson JF, Gutteridge E, Ellis IO, Pinder SE, Rubini M,
Nicholson RI: Epidermal growth factor receptor/HER2/insu-
lin-like growth factor receptor signalling and oestrogen
receptor activity in clinical breast cancer. Endocr Relat Cancer
2005, 12(Suppl 1):S99-S111.
37.
Knowlden JM, Hutcheson IR, Jones HE, Madden T, Gee JM, Harper
ME, Barrow D, Wakeling AE, Nicholson RI: Elevated levels of epi-
dermal growth factor receptor/c-erbB2 heterodimers medi-
ate an autocrine growth regulatory pathway in tamoxifen-
resistant MCF-7 cells. Endocrinology 2003, 144(3):1032-1044.
38.
Nahta R, Takahashi T, Ueno NT, Hung MC, Esteva FJ: P27(kip1)
down-regulation is associated with trastuzumab resistance
in breast cancer cells. Cancer Res 2004, 64(11):3981-3986.
39.
Amit I, Wides R, Yarden Y: Evolvable signaling networks of
receptor tyrosine kinases: relevance of robustness to malig-
nancy and to cancer therapy. Mol Syst Biol 2007, 3:151.
40.
Benjamini Y, Hochberg Y: Controlling the false discovery rate –
A practical and powerful approach to multiple testing. Journal
of the Royal Statistical Society Series B-Methodological 1995,
57(1):289-300.
41.
Tresch A, Beissbarth T, Sultmann H, Kuner R, Poustka A, Buness A:
Discrimination of direct and indirect interactions in a net-
work of regulatory effects. J Comput Biol 2007, 14(9):1217-1228.
|
19118495
|
ERa = ( MEK1 ) OR ( Akt1 )
p21 = ( ( ( ( ERa ) AND NOT ( Akt1 ) ) AND NOT ( CDK4 ) ) AND NOT ( cMYC ) )
CDK6 = ( CycD1 )
ErbB3 = ( EGF )
ErbB2_3 = ( ErbB2 AND ( ( ( ErbB3 ) ) ) )
pRB = ( CDK2 AND ( ( ( CDK4 AND CDK6 ) ) ) ) OR ( CDK4 AND ( ( ( CDK6 ) ) ) )
CDK2 = ( ( ( CycE1 ) AND NOT ( p21 ) ) AND NOT ( p27 ) )
ErbB2 = ( EGF )
Akt1 = ( ErbB1_3 ) OR ( ErbB2_3 ) OR ( ErbB1_2 ) OR ( ErbB1 ) OR ( IGF1R )
CycD1 = ( Akt1 AND ( ( ( ERa ) ) AND ( ( cMYC ) ) ) ) OR ( MEK1 AND ( ( ( cMYC ) ) AND ( ( ERa ) ) ) )
MEK1 = ( ErbB1_3 ) OR ( ErbB2_3 ) OR ( ErbB1_2 ) OR ( ErbB1 ) OR ( IGF1R )
IGF1R = ( ( Akt1 ) AND NOT ( ErbB2_3 ) ) OR ( ( ERa ) AND NOT ( ErbB2_3 ) )
ErbB1_2 = ( ErbB1 AND ( ( ( ErbB2 ) ) ) )
CycE1 = ( cMYC )
cMYC = ( Akt1 ) OR ( ERa ) OR ( MEK1 )
CDK4 = ( ( ( CycD1 ) AND NOT ( p27 ) ) AND NOT ( p21 ) )
p27 = ( ( ( ( ( ERa ) AND NOT ( CDK4 ) ) AND NOT ( cMYC ) ) AND NOT ( CDK2 ) ) AND NOT ( Akt1 ) )
ErbB1 = ( EGF )
ErbB1_3 = ( ErbB1 AND ( ( ( ErbB3 ) ) ) )
|
BMC Systems Biology
Research article
Reconstruction and logical modeling of glucose repression
signaling pathways in Saccharomyces cerevisiae
Tobias S Christensen†1,2, Ana Paula Oliveira†1,3 and Jens Nielsen*1,4
Address: 1Center for Microbial Biotechnology, Department of Systems Biology, Technical University of Denmark, Building 223, DK-2800 Kgs.
Lyngby, Denmark, 2Current address: Department of Chemical Engineering, Massachusetts Institute of Technology, Building 66, 25 Ames Street,
Cambridge, MA 02139, USA, 3Current address: Institute for Molecular Systems Biology, ETH Zurich, CH-8093, Zurich, Switzerland and 4Current
address: Department of Chemical and Biological Engineering, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
E-mail: Tobias S Christensen - tschri@mit.edu; Ana Paula Oliveira - oliveira@imsb.biol.ethz.ch; Jens Nielsen* - nielsenj@chalmers.se
*Corresponding author
†Equal contributors
Published: 14 January 2009
Received: 9 September 2008
BMC Systems Biology 2009, 3:7
doi: 10.1186/1752-0509-3-7
Accepted: 14 January 2009
This article is available from: http://www.biomedcentral.com/1752-0509/3/7
© 2009 Christensen et al; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Background: In the yeast Saccharomyces cerevisiae, the presence of high levels of glucose leads to
an array of down-regulatory effects known as glucose repression. This process is complex due to
the presence of feedback loops and crosstalk between different pathways, complicating the use of
intuitive approaches to analyze the system.
Results: We established a logical model of yeast glucose repression, formalized as a hypergraph.
The model was constructed based on verified regulatory interactions and it includes 50 gene
transcripts, 22 proteins, 5 metabolites and 118 hyperedges. We computed the logical steady states
of all nodes in the network in order to simulate wildtype and deletion mutant responses to
different sugar availabilities. Evaluation of the model predictive power was achieved by comparing
changes in the logical state of gene nodes with transcriptome data. Overall, we observed 71% true
predictions, and analyzed sources of errors and discrepancies for the remaining.
Conclusion: Though the binary nature of logical (Boolean) models entails inherent limitations,
our model constitutes a primary tool for storing regulatory knowledge, searching for incoherencies
in hypotheses and evaluating the effect of deleting regulatory elements involved in glucose
repression.
Background
Signaling and regulatory cascades establish the bridge
between environmental stimuli and cellular responses,
and represent a key aspect of cellular adaptation to
different environmental conditions. Cells can sense
several stimuli, both internally and externally, and the
received information will subsequently be propagated
through a cascade of physico-chemical signals. The
ultimate recipients of these signals will determine how
the cell responds, by acting at different regulatory levels
(transcriptionally, translationally, post-translationally,
allosterically, etc). Contrary to metabolic networks,
most signaling and regulatory pathways are relatively
poorly studied and signaling properties of a protein
cannot be easily derived from its gene sequence [1, 2].
Moreover, signal transduction networks operate over a
wide range of time-scales, and due to the presence of
feedback loops and cross talk it is difficult to discern
how concurrent signals are processed. Thus, methods to
analyze and model signal transduction and regulatory
circuits are of prime importance in biology, medicine
and cell engineering, since they can bring insights into
Page 1 of 15
(page number not for citation purposes)
BioMed Central
Open Access
the mechanistic events underlying complex cellular
behavior. The availability of models for signaling and
regulatory cascades represents an opportunity to expand
the search space when looking for intervention targets
that may lead to desired phenotypes – e.g. when looking
for better drug targets in medicine, designing novel
regulatory circuits in synthetic biology or finding
regulatory targets that can release metabolic control in
metabolic engineering.
Most eukaryotic cells, including many yeasts and
humans, can sense the availability of carbon sources in
their surroundings and, in the presence of their favorite
sugar (often glucose), trigger a cascade of signals that
will repress the utilization of less-favorite sugars as well
as the function of different catabolic routes [3-5]. This
phenomenon is commonly termed carbon-catabolite or
glucose repression. Because of its role in nutrient sensing
and its industrial impact on the simultaneous utilization
of different carbon sources, glucose repression has been a
model system for studying signaling and regulation. In
particular, glucose repression in the yeast Saccharomyces
cerevisiae has been extensively studied and two main
signaling pathways have been identified: a repression
pathway, mediated through the protein kinase Snf1 and
the transcription factor Mig1, and a glucose induction
pathway, mediated through the membrane receptors
Snf3 and Rgt2 and the transcription factor Rgt1 (for
review see, for example, [4, 6-8]). Growing evidence
suggests the existence of extensive cross talking between
these two pathways. Figure 1 summarizes key aspects of
the system. Besides the role of glucose repression on the
utilization of alternative carbon sources, glucose repres-
sion in yeast leads to the transcriptional shutdown of
genes related to respiration, mitochondrial activities, and
gluconeogenesis [9]. This transcriptional behavior causes
the wild-type yeast to exert respiro-fermentative meta-
bolism during growth on excess glucose, redirecting
carbon towards by-products of metabolism such as
ethanol, acetate and glycerol, at the cost of biomass
formation. Despite being an extensively studied system,
knowledge on yeast glucose repression is still far from
complete and key questions remain, including: what
exactly triggers the cascade signal(s)? How to differenti-
ate between causes and consequences? How does the
knowledge derived from phenotypic observations relate
to mechanistic events? How does the current knowledge
on glucose repression fit with available high-throughput
data? In order to attempt to bring insights into these
questions, we aim here at creating a mechanistic, semi-
quantitative model of glucose repression signaling
cascades and genetic regulatory circuits in yeast.
Modeling approaches of different levels of abstraction
have been proposed to analyze and simulate signal
transduction and regulatory networks, ranging from
purely topological to kinetic models. While attractive
in principle, quantitative kinetic models based on ODEs
are hampered by difficulties in determining the necessary
parameters and kinetic equations. At the other extreme,
strictly descriptive models have also been reported
[10-12], in which the precision of the formalism
proposed, based on process engineering, establish a
clear and unique qualitative representation of the
network interactions. Somewhere in between lie more
semi-quantitative and qualitative approaches that
require the topological description of signaling interac-
tions and make use of well-established mathematical
frameworks to analyze network structure and function-
ality. Such methods include (i) stoichiometric represen-
tation and extreme pathway analysis [13], (ii) Boolean
(on/off) and Bayesian (probabilistic) representation of
interactions [14-16], (iii) logical hypergraph representa-
tion and logical steady state analyses [17], and (iv) Petri
nets graph representation and simulations [18-20]. All
these methods describe signal flow qualitatively within
a mathematical formalism and without loss of
Figure 1
Simplified representation of the main glucose
repression pathways. Glucose is transported into the cell
by hexose transporters with different affinities (HXT1-
HXT16). Inside the cell, glucose is phosphorylated to glucose
6-phosphate by Hxk2, therefore entering into carbon
metabolism. An unknown signal triggered by high glucose
levels leads to inactivation of the Snf1 complex. This
inactivation is regulated by the protein phosphatase Glc7-
Reg1. Inactive Snf1 cannot phosphorylate Mig1, which thus
remains in the nucleus under high glucose levels, exerting
repression of transcription of several genes. At low glucose
concentrations, when Snf1 becomes active, Mig1 is
phosphorylated and translocates to the cytosol, releasing
repression. Glucose is sensed by two sensors located in the
cell membrane, Rgt2 and Snf3. At high glucose levels, the
signal from these sensors leads to SCFGrr1 mediated
ubiquitination and consequent degradation of Mth1 and Std1,
which are required for Rgt1 activation.
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 2 of 15
(page number not for citation purposes)
information on the network topology, allowing insight-
ful computations on network structure such as evaluat-
ing the degree of cross talk, determining all possible
elementary 'flux' modes and calculating the number of
theoretically possible positive and negative feedback
loops. Moreover, logical hypergraph analyses and Petri
net models also have the potential to be used for semi-
quantitative simulation of network behavior, since they
allow simple predictions of the state of a system in
response to different stimuli.
In this work, we reconstructed the signaling and
transcriptional regulatory network of glucose repression
in S. cerevisiae based on established knowledge reported in
the literature. We converted this information into a
logical hypergraph, and performed structural and func-
tional analyses on the network following the framework
proposed by Klamt and co-workers [17]. Next, we
performed logical steady state analyses to compute the
state of all nodes in the system under all possible
environmental conditions (sugars availability), and for
all different single gene deletions and some double gene
deletions. Furthermore, we developed a framework to
evaluate model predictions by comparing changes in the
state of the regulatory layer against changes in gene
expression data (transcriptome data was available for
several knockouts of the system). Based on the results
from the model evaluation, we identified main errors
and discuss possible sources of discrepancies, as well as
the inherent limitations to Boolean modeling. Our
results point towards the existence of incoherencies
between high-throughput data and literature-based
knowledge related with glucose repression. To our
knowledge, this represents the first attempt to mechan-
istically and semi-quantitatively model glucose repres-
sion signaling and regulatory pathways in the yeast S.
cerevisiae.
Results
Reconstruction of the signaling/regulatory network
and model setup
Glucose repression signaling and regulatory network was
reconstructed from low-throughput data reported in
peer-reviewed publications. Information was gathered
based on biochemical studies and physiological obser-
vations, and it was included in a collection database
containing: (i) list of proteins with sensor, signaling or
transcription factor functions found to be related to
glucose repression; (ii) list of genes known to be
transcriptionally regulated by glucose repression related
transcription factors; (iii) type of regulation exerted on
each of the previous species by metabolites and/or
regulatory proteins. The reconstructed network accounts
for 72 species (corresponding to 50 genes) and 148
interactions, which cover most of the current knowledge
on the Mig1/Snf1 and Snf3/Rgt2 pathways, as well as
galactose and maltose regulatory systems. Transcription
factors included are Rgt1, Mig1, Mig2, Mig3, Sip4, Cat8,
MalR and Gal4. Regulatory targets include genes encod-
ing hexose transporters and enzymes involved in maltose
catabolism, gluconeogenesis and the Leloir pathway. The
complete list of species and interactions considered is
given as supplementary material (Additional file 1).
Thereafter, the reconstructed signaling/regulatory net-
work was converted into a logical hypergraph (Figure 2),
representing all interactions in a logical manner (Figure 3
and Additional file 2), according to the framework
proposed by Klamt et al. [17] to model signaling
networks. The conversion of signaling and regulatory
interactions into Boolean functions was based on
described functions reported in the literature (the
rationale for the choice of less obvious Boolean
functions for certain interactions is explained in the
Additional file 2). The resulting hypergraph consists of
77 nodes (50 genes, 22 proteins, 3 extracellular
metabolites and 2 intracellular metabolites) and 118
hyperedges, and represents a logical model for glucose
repression signaling and regulatory pathways. For ease of
visualization, we have depicted the hypegraph into four
separate sub-networks, each representing in more detail
a different system: Mig1/Snf1 and Snf3/Rgt2 pathways
(Figure 3A), galactose regulation (Figure 3B), maltose
regulation (Figure 3C) and the Sip4/Cat8 regulatory
system (Figure 3D). The model takes as input the
availability of carbon sources (glucose, galactose, and
maltose) and outputs the logical steady state of the
network.
In our model, nodes can assume one of two logical
states, 1 or 0, corresponding to on or off, or in more
subtle instances, higher or lower activity. For protein
nodes, this can most simply be interpreted as a protein
being active (1) or inactive (0), whereas in the case of
gene nodes, it can be seen as a gene being expressed (1)
or not (0). We used the model to analyze structural
characteristics of the network and to compute logical
steady states of all nodes in the network. In particular,
we simulated how gene transcripts change their logical
state in response to perturbations (e.g., availability of
different sugars and different gene knockouts), and
evaluated the predictions by using available transcrip-
tional datasets for different carbon source conditions
and yeast deletion mutants. During logical states
simulations, although most nodes were left uncon-
strained, a few nodes were assigned a default value of 1.
E.g., GRR1 was set to a fix state of 1 since its regulation is
not considered in the model (otherwise, no unique
logical steady state would exist). Also, in the case of
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 3 of 15
(page number not for citation purposes)
genes constitutively expressed at a basal level such as
MALT, encoding a maltose transporter, the node state
was set to a fix value of 1.
Structural and functional analyses of the network
Logical steady state analyses were performed for all
combinations of sugar availability for the wild type, all
single gene knockout mutants (24 cases), and three
double gene deletion mutants of interest, in a total of
224 simulations (see Additional file 3). We notice that
most of the gene nodes change their logical state in over
Figure 2
Example of Boolean expressions and corresponding
logical hypergraphs and truth tables. A logical
hypegraph is an interaction network where each edge (or
hyperarc) connects a set of start-nodes (tails) to an end-node
(head), and the combination of incoming hyperarcs to an
end-node represents a Boolean expression. Any Boolean
expression can be written in a disjunctive normal form (only
using AND, OR and NOT operators). On the disjunctive
normal form, expressions are built up by literals (i.e.
variables or their negation) connected by AND relations
forming clauses, and clauses can then be connected by OR
relationships. In the logical hypergraph, each clause is
represented by a hyperarc, while separate hyperarcs linked
to an end-node represents clauses connected by an OR
relationship. When the value of a tail species is negated, this
is marked by a repression symbol in the corresponding
hyperarc (see also the symbolic explanation in Figure 3). The
Boolean function determining the state of a node is thus
given by all the incoming hyperedges.
Figure 3
Hypergraph representation of the Boolean model for
yeast glucose repression. A) Overall representation of
the main glucose repression pathways, including the Snf1/
Mig1 repression pathway and the Snf3/Rgt2 induction
pathway. Note how the high degree of cross talk makes it
impossible to distinguish between the two pathways. B) The
GAL regulatory system. C) The MAL regulatory system.
MALT is set to be active by default, since MalT is assumed to
be present at a basal level. D) Subset of the network
controlled by Sip4 and Cat8. Signaling proteins are
represented with purple rectangles, while transcription
factors are represented with red rectangles. Accordingly, a
post-transcriptional regulatory association is represented
with a purple line, while a transcriptional regulatory
interaction is represented with a red line. Gene transcripts
are represented with green rectangles, and sugar metabolites
are depicted by blue ellipses. The description of the
representation of logical relations between species is given in
the legend of Figure 2. A note about the nomenclature in the
hypergraph representation: genes are denoted in uppercase
(e.g., MIG1), while protein names include the suffix 'p' (e.g.,
Mig1p). In the main text, proteins are named similarly, but
without the suffix (e.g., Mig1), and genes are in italic
uppercase (e.g., MIG1)
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 4 of 15
(page number not for citation purposes)
10% of the simulations, but only few (MTH1, MALR,
GAL3, GAL4 and CAT8) change in more than 15% of the
simulations. The predictions for the wild type along with
a subset of the deletion mutants for which transcriptome
data was available were analyzed and used to evaluate
the model, as will be discussed below. The capability to
make semi-quantitative predictions of gene expression
levels and protein activity for any combination of gene
deletions and nutrient conditions is a key feature of the
model.
The reconstructed network contains 35 negative and 14
positive feedback loops, which is indicative of the high
degree of crosstalk between pathways and complex
cause-effect relationships. This is further supported by
the dependency matrix of the network (Figure 4), which
is based solely on the underlying network interaction
graph without information on Boolean functions. In the
matrix, yellow elements represent an ambivalent rela-
tionship between an ordered pair of species (i,j), where i
and j represents the column and the row number,
respectively, in the sense that both activating and
inhibiting paths exist from i to j. Dark or light green
(/red) elements, Dij, indicate that species i is a total or
non-total activator (/inhibitor) of species j, respectively,
i.e. only activating (/inhibiting) paths from i to j exist
and feedback loops are either absent (total) or present
(non-total) – see Figure 4 legend for details. Examining
the dependency matrix, the large number of ambivalent
relationships (represented by yellow elements) as well as
the prevalence of negative feedback loops in signaling
paths (light green or red fields) is noteworthy. It
underscores the difficulties in making predictions
based on intuitive approaches and emphasizes the
need for a logical modeling framework. The matrix
also reveals the high degree of crosstalk between the
Mig1/Snf1 and Snf3/Rgt2 pathways, since it is quickly
noted that the signaling proteins in one pathway
generally affect proteins in the other pathway.
Model evaluation
In order to evaluate the capability of the logical model to
predict differential gene expression, we performed
logical steady state analysis of the glucose repression
regulatory response for five different gene knockouts,
and compared the results with available whole-genome
gene expression from DNA-microarrays. We used data
from the yeast mutants Δrgt1, Δmig1, Δmig1Δmig2,
Δsnf1Δsnf4, Δgrr1 and their isogenic reference strains.
This type of analysis not only gives an indication of the
model's predictive strength, but also hints at possible
errors in the model (and eventually in the underlying
hypotheses from the literature) in the cases where
discrepancies between model and observation occur.
Figure 4
The dependency matrix for the yeast glucose
repression network. Each element in the matrix shows the
relationship between an effecting species and an affected
species, specified at the bottom of the column and at the end
of the row, respectively. A yellow field in the intersection of
the ith column and jth row signifies that the ith species is an
ambivalent factor with respect to the jth species, i.e. that
both activating and repressing/inhibiting paths from the ith to
the jth species exist. For example, the yellow color of the first
element (first column and first row) indicates that both
inhibiting and activating paths exists from exterior glucose to
the Sip4 protein, i.e., exterior glucose is an ambivalent factor
with respect to Sip4. Similarly, a dark green field and a light
green field indicate a total and a non-total activator,
respectively, i.e., only activating paths exist and negative
feedback loops are either absent (total) or present (non-
total). Dark red and light red fields represent total and non-
total repressors/inhibitors, respectively. A black field
indicates that no path exists from A to B. The large number
of black columns in the middle corresponds to output
species (sinks), which per definition are non-affecting
towards all species. Due to the directional nature of the
interaction network, the matrix is not symmetric (e.g. Sip4 is
non-affecting toward exterior glucose). See nomenclature
note for gene and protein species in the legend of Figure 3.
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 5 of 15
(page number not for citation purposes)
The best we can hope to achieve with a Boolean model is
a correct prediction of the sign of the change in gene
expression, i.e., the model prediction Yi
mod should equal
the experimental observation Yi
exp when evaluating
change in expression of gene i following a knockout or
change in conditions. In order to assess what experi-
mental results should be regarded as a change, it is
necessary to make an interpretation of the gene expres-
sion data that allows a comparison with the binary
outputs of the Boolean model. Intuitively, the experi-
mental change should be relatively large and statistically
significant in order to be reflected by the discrete
Boolean model. Therefore, we established a fold-change
threshold ([FCmin| = 1.5; see Methods for fold-change
definition) and a Student's t-test p-value cut-off (a =
0.05) for all pairwise gene expression comparisons
between a deletion mutant and its isogenic reference
strain. All genes with p-value < a and FC ≥FCmin (or FC ≤
-FCmin) were assigned with a value of Yi
exp = 1 (or Yi
exp =
-1), and 0 otherwise. Such conversion of gene expression
into discrete Boolean values based on a somewhat
subjective threshold may yield a number of type-2 (false
negatives) and type-1 (false positives) errors.
The hereby identified experimental variation, Yi
exp, was
then compared with the model prediction, Yi
mod, for
each gene i. Yi
mod was determined by the difference in
Boolean output for gene i between the mutant and the
reference state (wildtype), at a defined external condi-
tion. Thus, Yi
mod can assume the values -1, 0 or 1,
corresponding to a decrease, no change or increase in
gene expression on transcript i in the mutant, respec-
tively. Model prediction capabilities were evaluated
based on the difference |Yi
mod - Yi
exp|, with a value of
0 meaning a correct prediction, a value of 1 implying a
small error, and a value of 2 indicating a large error (only
happening when model prediction and experimental
results point towards opposite directions).
A summary of the results from the comparison between
model prediction and experimental up- and down-
regulation for all five different knockouts evaluated is
shown in Table 1. In the following, we discuss more
thoroughly the results for Δrgt1, and use this to analyze
common reasons for discrepancies in all the knockouts.
The remaining comparisons are briefly commented
afterwards.
Evaluation of Δrgt1 mutant
Transcriptome data for the yeast Δrgt1 mutant and its
isogenic reference strain is available from [21] during
shake flasks cultivations using galactose as the single
carbon source. Sampling was made in the mid-exponen-
tial phase, where pseudo-steady-state can be assumed
(i.e., growth rate and physiological yields appear
constant during the exponential phase, despite changes
in the concentration of extracellular metabolites). We
converted the gene expression data from this study into
Yi
exp according to the procedure described above, and
compared it with the simulation results Yi
mod (Table 1).
Yi
mod are derived from logical steady state analyses of the
logical model assuming galactose present and all other
carbon sources absent. The logical steady states were first
determined for the original model without further
constrains (Xi,WT). Afterwards, in order to simulate the
RGT1 gene deletion, the node RGT1 of the hypergraph
was set to zero, and a new logical steady state analysis
was performed (Xi,RGT1). Yi
mod is given by the difference
between Xi,RGT1 and Xi,WT.
In general, the model predictions are very good for the
Δrgt1 mutant, with 82% true predictions, reflecting the
fact that Rgt1 along with its regulators has been
extensively studied [21-30]. Only for 6 out of the 34
genes evaluated, the experimental fold changes do not
correspond to model prediction. The six genes are SNF3,
MTH1, MIG2, MAL33 (which encodes a MAL regulator),
SUC2 and HXT8. In the following, the causes of these
discrepancies are investigated.
Analysis of discrepancies in SNF3. SNF3 encodes a high-
affinity glucose sensor located in the plasma membrane.
Gene expression data shows no differential expression of
SNF3 in the Δrgt1 mutant (p-value = 0.88, FC = 1.03).
Table 1: Summary of results from the model evaluation
Knockout Genotype
Number of Genes
Evaluated
Number of true
predictions
Percentage of true
predictions for
genes changed on
array
Percentage of true
predictions for
genes changed in
the model
Percentage of true
predictions
Δrgt1
34 (25)
28 (21)
83% (83%)
71% (63%)
82% (84%)
Δmig1
38 (27)
24 (20)
29% (44%)
40% (57%)
63% (74%)
Δmig1Δmig2
38 (28)
24 (19)
47% (53%)
69% (80%)
63% (68%)
Δsnf1Δsnf4
34 (25)
17 (14)
45% (60%)
56% (64%)
50% (56%)
Δgrr1
38 (28)
15 (10)
25% (24%)
55% (100%)
39% (43%)
In parentheses are the numbers when dubious genes are not included in the computations (see Discussion for details).
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 6 of 15
(page number not for citation purposes)
However, in the model this gene is found to be down-
regulated in the mutant relative to the wild type. The
most likely explanation for this discrepancy lies in the
logical equation for SNF3 applied in the model:
SNF3 = NOT (Mig1 OR Mig2)
= (NOT Mig1) AND (NOT Mig2)
In the model, RGT1 deletion leads to an active Mig2 and
consequently repression of SNF3. The most plausible
explanation for this is that the model overestimates the
importance of Mig2 and that, in reality, the presence of
active Mig2 by itself is not enough to prevent SNF3
transcription. Nevertheless, this explanation cannot be
accepted out of hand: the dependency matrix reveals that
RGT1 is an ambivalent factor with regard to SNF3, i.e.,
both repressing and activating paths from RGT1 to SNF3
exist. It is thus possible that Mig2 is indeed a significant
repressor of SNF3, but that other important, repressing
pathways from RGT1 to SNF3 are also deactivated by the
RGT1 deletion.
A closer inspection of the signaling paths from RGT1 to
SNF3 in the hypergraph reveals that, from the 8 existing
possible paths, 4 are repressing paths of the form RGT1 -
> Rgt1 -| Mig2/Mig3 -| Mig1 -| SNF3. This means that
Rgt1 represses Mig2 and Mig3, both of which repress
Mig1, which then represses SNF3 (this constitutes 4
paths because two hyperedges connect RGT1 to Rgt1,
either with Mth1 or Std1 as the second tail). It seems
unlikely that these 4 signal flux modes play significant
physiological roles since (i) no significant role of Mig3
has ever been found [31-33], and (ii) Mig2 can at most
serve to attenuate Mig1 expression since the two proteins
are active at basically the same conditions.
Analysis of discrepancies in MTH1. Mth1 is a signaling
protein intermediate between the membrane sensors
Snf3/Rgt2 and the transcription factor Rgt1 [23, 24].
Gene expression of MTH1 is found to be up-regulated in
the Δrgt1 mutant, whereas the model predicts the
expression level of MTH1 to be unchanged. Despite
the fact that Rgt1 repression of MTH1 is reported in the
literature (in fact in the same paper where Δrgt1
transcriptome data is presented) [21], the repression of
MTH1 by Rgt1 is ignored for logical steady state
calculations for two reasons. Firstly, Kaniak et al. state
that this transcriptional repression is weak (the tran-
scriptome analysis was actually complemented with
promotor-lacZ fusions, and it was found that MTH1,
both in the wild-type and in the Δrgt1 mutant, is subject
to considerable glucose repression, not glucose induc-
tion as would be expected for a gene primarily regulated
by Rgt1 [21]. Secondly, in terms of the Boolean model,
including Rgt1 in a "NOT Rgt1 AND-relationship" would
mean that Mth1 would always be inactive when Rgt1 is
active, which would be somewhat incongruous consider-
ing that MTH1 seems to encode a co-repressor of Rgt1.
This example illustrates the difficulties in incorporating
negative feedback loops in a binary Boolean model.
It should also be noted that MTH1 is, according to the
model at least, one of the most heavily regulated genes
in the network, being transcriptionally regulated by Rgt1,
Mig1, Mig2, Mig3 and Gal4. This makes a literature
based determination of the Boolean function governing
its expression particularly difficult.
Analysis of discrepancies in MIG2 and HXT8. MIG2
encodes for a homologue of the transcription factor
Mig1, while HXT8 encodes for a plasma membrane
hexose transporter. MIG2 was not found to change
experimentally, at least not in terms of our defined
"Boolean fold change" threshold, but was found to be
up-regulated in the model. This discrepancy is, most
likely, due to a type-2 error in the inference of gene
expression change from the transcriptome data. Even
though the average fold change observed experimentally
was 3.4, the p-value of this change was only 0.29. As this
is above the cut-off value of 0.05, this change is deemed
insignificant and the gene is attributed a "Boolean fold
change" of 0. Nevertheless, the model prediction is in
good agreement with the results of the continued
investigation by Kaniak et al., which included promo-
ter-lacZ fusions and ChIP experiments, and showed that
Rgt1 is a strong (and possibly the only) transcriptional
repressor of MIG2. The discrepancy for HXT8 seems to
have similar reasons.
Analysis of discrepancies in MAL33 (MALR). Expression of
MAL33, encoding a MAL regulator, was found to be
experimentally repressed in the mutant, while it
remained unchanged in the model. The three genes
required for maltose metabolism are mapped in various
MAL loci, of which 5 are currently known [34, 35]. In the
model, no distinction is made between the different
complex loci. Since this is a recurrent discrepancy in all
evaluations performed, we present a general discussion
on the MAL regulatory system later in the Discussion
section.
Analysis of discrepancies in SUC2. Expression of SUC2,
encoding for invertase (sucrose hydrolyzing enzyme),
was not found to be significantly different between the
Δrgt1 mutant and wild-type, whereas the model predicts
a decrease in gene expression in the mutant. This
discrepancy illustrates both the difficulties in choosing
the correct logical equation for a specific species based
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 7 of 15
(page number not for citation purposes)
on literature and in converting a continuous reality to a
binary model.
In the model it is assumed that both Mig1 and Mig2
should be absent in order for SUC2 to be expressed at
high levels (SUC2 = NOT (Mig1 OR Mig2)), cf. Lutfiyya
et al. who found that the single deletions had relatively
low impact on SUC2 expression level, whereas Δmig1Δ-
mig2 double deletion had great effect [36]. Contrary to
this, Klein et al. found a large increase in SUC2
expression in a Δmig1 strain and further increase by
additional disruption of MIG2 [37]. The Klein et al.
observations could have been implemented in the model
via a Boolean function where the absence of either one
of the two repressors induces expression of SUC2 (i.e.,
SUC2 = NOT (Mig1 AND Mig2)) or, alternatively, by
simply ignoring Mig2 in the equations. While imple-
menting either of the alternatives would have led to
correct model prediction for the knockout (as judged by
comparison with the expression data), the actual output
for SUC2 in the wild-type would have been the same
regardless of the chosen equation (since absence of Mig
1 or of both Mig1 and Mig2 would always result in active
SUC2). Nevertheless, the ambiguity in the literature
combined with the difficulty of imposing a discrete
model on a continuous reality made the choice of logical
equation extremely hard in this case.
The model apparently mistakenly predicts a decrease in
SUC2 expression, because RGT1 deletion causes MIG2
to be expressed (i.e. Mig2 becomes active) which then
leads to repression of SUC2 with the chosen Boolean
equations. Based on this evaluation, and assuming that a
type-2 error has not occurred, it therefore seems reason-
able to say that the model overestimates the importance
of Mig2 in the regulation of SUC2. Alternatively, it is
possible that MIG2 transcription is induced, but that the
Mig2 protein is also post-translationally regulated, some-
thing that is not described in the model or, to the best of
our knowledge, in the literature. This could mean that
Mig2 protein activity is not increased by the deletion of
RGT1, despite the eventual increase in transcript levels.
Evaluation of Δmig1 and Δmig1Δmig2 mutants
Transcriptome data for Δmig1 and Δmig1Δmig2 mutants,
and their isogenic reference strain was available from
[38] during aerobic batch cultivations using 40 g/L
glucose as single carbon source. Samples were taken in
the mid-exponential phase at a residual glucose con-
centration of 20 g/L. Therefore, experimental observa-
tions were evaluated against model predictions with only
glucose present. For both knockouts, the percentage of
true predictions is of 63% (Table 1). When examining
the discrepancies between experimental observations
and model predictions, it is particularly noticeable that
model predictions for MAL and GAL regulatory genes are
almost always wrong. In particular, in the Δmig1 case
both MAL13 and GAL4 evaluation produces a large error
(i.e., |Yi
mod - Yi
exp| = 2), meaning that the experimental
direction of regulation is opposite to that predicted by
the model. Whereas in the case of MAL genes this is one
of several discrepancies observed in this work (see
Discussion), in the case of GAL4 we find the experi-
mental gene expression data from [38] to directly
contradict the results of a Northern blot analysis [39].
While the former study showed a decrease in GAL4 levels
upon deletion of MIG1, the second showed more than 6-
fold increase in the levels of GAL4 transcripts in a Δmig1
mutant, in a medium with the same glucose concentra-
tion (as cited in [40]).
Evaluation of Δgrr1 mutant
Transcriptome data for Δgrr1 and its isogenic reference
strain were also available from [38], at conditions
corresponding to high extracellular glucose concentra-
tion. Evaluation of model predictions for Δgrr1 yielded
the poorest results, with only 39% of true predictions
(Table 1). This is probably due to the fact that, despite
being an important player in a large number of cellular
activities such as cell morphology, heavy metal tolerance,
osmotic stress and nitrogen starvation [25, 30], Grr1 is in
the model only represented as a simple regulator acting
upstream of Rgt1. The highly pleiotropic nature of Grr1
is revealed in the DNA-microarray data, where 24 out of
38 evaluated genes are found to be differentially
expressed (in the Boolean sense). The low percentage
of correct predictions illustrates the danger of including
species without properly accounting for all key func-
tions. We notice that the model even fails to efficiently
predict the alteration of gene expression for targets of
Rgt1 (e.g. HXT2, HXT4, HXT5 and HXT8), which,
according to the model, should act downstream of
Grr1. However, given the scale of the perturbations
caused by GRR1 deletion, it is difficult to say whether
these discrepancies are caused by faults in the current
hypotheses underlying the model or by secondary effects
not
taken
into
consideration,
such
as
altered
metabolism.
Evaluation of Δsnf1Δsnf4 mutant
Transcriptome data for Δsnf1Δsnf4 and its isogenic
reference strain were available from [41], during aerobic
continuous cultivations at dilution rate 0.1 h-1 and
glucose (10 g/L) as single carbon source. Under these
conditions, the residual glucose concentration inside the
fermentor is very low, and typically no glucose repres-
sion is observed. This behavior is, to some extent, similar
to what happens in the absence of glucose. Thus,
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 8 of 15
(page number not for citation purposes)
experimental observations were compared with model
predictions for the case where all sugars were absent. The
percentage of true predictions was only 50% (Table 1). A
surprising discrepancy was observed for the expression
levels of CAT8 and SIP4, which were predicted to
decrease, but are found to be experimentally unchanged
(in the Boolean sense). Nevertheless, the prediction of a
down-regulatory effect on Cat8 and Sip4 gene targets
(ICL1, FBP1, PCK1, MLS1, MDH2, ACS1, SFC1, CAT2,
IDP2 and JEN1) is confirmed experimentally. While the
observed changes for CAT8 and SIP4 are not statistically
significant, they are nevertheless in the predicted direc-
tion, and the occurrence of type-2 errors is therefore
likely. An additional and likely explanation is that the
effect of the Snf1-Snf4 complex on repression by Sip4
and Cat8 is, to a larger extent, mediated via direct
posttranslational regulation rather than via indirect,
Mig1 mediated transcriptional regulation.
Global evaluation of predictive power
Boolean models lie at the boundary between qualitative
and quantitative models. For the present model of
glucose repression, testing current hypotheses is at least
as important as making predictions. These goals, how-
ever, are connected in the sense that one way in which
inconsistencies in the model (and consequently, in the
hypotheses proposed in the literature) are revealed is
through failure to make predictions. Therefore, evaluat-
ing the predictive power of the model is an important
task. Above, we looked into the capacity of the model to
predict differential gene expression for individual knock-
outs, and observed that the frequencies of correct
predictions varied between the different knockouts
(and in no case were all predictions true). What remains
to be demonstrated is that the correct predictions were
not obtained by chance.
For the simplest random model, the probabilities that
the expression of a gene is predicted to be unchanged,
up-regulated or down-regulated, respectively, would all
equal 1/3. Overall, the average probability of having the
correct prediction in each situation would also be 1/3.
Table 1 shows that, for all knockouts, the frequency of
correct predictions exceeds 33%. We tested the signifi-
cance of this occurrence by applying a binomial
distribution test and p0 = 0.33 (cf. Methods) to all
predictions (Table 2). Overall and for most knockouts,
the very low p-values unambiguously show that the
results were not obtained due to chance. However, for
Δgrr1 it cannot be shown at 95% confidence level that
the prediction rate is significantly better than with a
random model.
Next, we applied the same distribution to test the
significance of the fraction of large errors observed
(i.e., |Yi
mod - Yi
exp| = 2) in the subset of cases where
differential expression was observed experimentally as
well as in the model. Since in this case there would be
50% chance of a correct prediction if nothing was known
a priori, we use p0 = 0.5. Out of 41 cases, only 3 cases of
opposite signs predictions are observed. This is signifi-
cant with a p-value less than 10-90. The three encountered
large errors were in two predictions for MALR (discussed
below) and one prediction for GAL4 in the Δmig1
mutant (most likely arising from the microarray experi-
ment itself, as discussed above).
Discussion
The goal of our Boolean network modeling and analyses
was dual. First, we wanted to use the model to test how
the underlying biological hypotheses (for signal trans-
duction and transcriptional regulation) found in litera-
ture fit the observations from genome-wide gene
expression studies of different signaling knockout
mutants. We proposed a framework to compare simula-
tion results with experimental observations and looked
for discrepancies between the two. These discrepancies
hinted at the identification of different types of errors, as
discussed below. Second, we wanted to evaluate the
model's predictive strength and investigate to what
extent we could use it to simulate transcriptional
responses upon deletion of various components of the
glucose repression cascade. Such a model can eventually
be combined with genome-scale stoichiometric meta-
bolic models to further constrain the solution space
during optimization problems (e.g., flux balance analy-
sis). Gene expression data in the form of discrete
Boolean on/off information has been previously used
to constrain fluxes (or more precisely, the genes
encoding the enzymes catalyzing the corresponding
Table 2: Statistical evaluation of the significance of the model prediction
H0: X/N = 1/3
H1: X/N > 1/3
Δrgt1
Δmig1
Δmig1Δmig2
Δsnf1Δsnf4
Δgrr1
Overall
p-value
6.7 × 10-10
4.8 × 10-5
4.8 × 10-5
1.2 × 10-2
2.1 × 10-1
2.6 × 10-14
P-values for the statistical evaluation of the null hypothesis, H0, tested for whether the observed level of success was due to chance, i.e. that the
average proportion of correct predictions (X/N) is 1/3, against the alternative hypothesis that the model yields a higher proportion of true
predictions. The test is based on the binomial distribution (cf. Methods).
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 9 of 15
(page number not for citation purposes)
reactions) in stoichiometric metabolic models, adding a
layer of transcriptional regulation into this type of
models [42, 43]. Our approach goes even further by
also including signaling information, opening new doors
to search for targets that can release metabolic control at
different regulatory levels.
Glucose repression is a complex and intertwined
regulatory system, with extensive cross-talk among
pathways, feedback loops and different levels of regula-
tion responding at different time scales. This makes it
difficult to decide the logical rules describing some of the
species and their influence, in particular species that are
heavily regulated (e.g., Mth1) or those with extensive
pleiotropic effects (e.g., Grr1). Noticeably, we explicitly
decided not to include hexokinase-2 (Hxk2) in the
model, despite Hxk2 being hinted to be a key regulator
in the Snf1/Mig1 pathway (in addition of its role as a
glycolytic enzyme). This decision was based on observa-
tions that changes in the activity of Hxk2 lead to an array
of effects, namely altered metabolism, which may
indirectly trigger other regulatory responses [38, 44].
Thus, it becomes difficult to describe the role of Hxk2 in
a model of moderate size like ours. More knowledge on
the exact signaling and regulatory roles of Hxk2 in
glucose repression will be necessary before it can be
included in our model.
We performed logical steady state analyses for the
wildtype, all single gene deletions and three double
gene deletions under all combinations of sugar avail-
ability, and observed that most nodes change their
logical steady state in more than 10% of the gene
deletion simulations. Thereafter, we evaluated the model
predictions against available gene expression data by
comparing changes in transcript levels (converted into a
Boolean form) with simulations of the logical steady
state of the model. Determination of the overall true
prediction rate for the analyzed knockouts shows that
Δrgt1 yielded the best results (82%), while true predic-
tions for Δgrr1 were the weakest (only 39%). The highest
prediction rate found for Δrgt1 is probably related with
the fact that many of the regulatory mechanisms
included in our model are originated from transcrip-
tional studies on the role of Rgt1. Conversely, the very
bad prediction capabilities for Δgrr1 are likely related
with the pleiotropic role of Grr1 in nutrient sensing, and
the fact that our model does not account for all the
regulatory effects associated with Grr1. Somewhere in
between, true predictions for Δmig1, Δmig1Δmig2 and
Δsnf1Δsnf4 lay in the range 50%–63%. These prediction
rates can be improved if we do not account for genes
with dubious regulation. Namely, the genes HXT5,
HXT8, YGL157W, YKR075C, YOR062C, YNL234W and
the MAL loci were included in the model even tough
little is known about their regulation. These genes are
presumably regulated by one or two of the transcription
factors in the model via a hypothesized mechanism (see
Additional file 1), but may very well also be regulated via
other mechanisms, possibly involving other regulators.
Nevertheless, their inclusion in the model allows us to
see to what extent their expression is explained by the
proposed mechanism. We observed that, if these
dubiously regulated genes were not included for model
evaluation, the rate of correct predictions increased
markedly in the case of Δmig1, Δmig1Δmig2 and
Δsnf1Δsnf4 (see values in parenthesis in Table 1).
Moreover, if we exclude the genes with dubious
regulation mentioned above as well as the results from
the highly pleiotropic knockout of GRR1, we observe an
improvement in the overall success rate from 60% to
71%. This suggests that the proposed mechanisms are
probably incomplete for these genes.
Overall, analyses of the discrepancies between model
predictions and transcriptome data hints at four main
sources of errors: (i) errors arising from imprecise
conversion of knowledge into logical representation,
(ii) errors inherent to the Boolean formalism, (iii) errors
arising from the discrete evaluation of experimental gene
expression data, and (iv) situations where high-through-
put data goes against literature-based knowledge.
Errors arising from imprecise conversion of knowledge into
logical representation. Many of the discrepancies found
in the model are originated from biological ambiguities
or from difficulties in translating biological behavior
into logical rules. For example, during evaluation of the
Δrgt1 results we saw that it was not trivial to describe in
Boolean terms the regulation of SNF3 and SUC2 by Mig1
and Mig2. In both cases, the error seems to arise from an
over-estimation of the importance of Mig2 repression.
Although Mig1 and Mig2 are believed to have somewhat
redundant roles as repressors, the Boolean formalism
does not make it easy to distinguish different levels of
regulation. Most notoriously, the model fails to predict
the response of the MAL genes (only 1/3 of the
predictions were correct, which is the same frequency
expected for a random model). In S. cerevisae the MAL
genes, that are required for utilization of maltose as
carbon source, co-locate in telomere-associated MAL
loci. Although there are five different MAL loci identified
[45] there is large variations in terms of presence and
activity of these loci in different strains [35]. Moreover,
experimental studies suggest that different MAL loci are
regulated by distinct regulatory mechanisms, and, in
particular, the MAL6 locus has been reported to be
regulated differently than the MAL1, MAL2 and MAL4
loci [45, 46]. In the present work, network reconstruction
of the MAL system was based on investigations of the
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 10 of 15
(page number not for citation purposes)
MAL6 locus [34, 45, 47]. However, during model
evaluation we made no distinction between different
alleles – e.g. MALR is used to represent all MAL activator
encoding genes, i.e. MAL13, MAL23, MAL33, MAL43 and
MAL63 (cf. [48]). This generalization may be a major
reason for the very low prediction rates.
Errors inherent to the Boolean formalism. Another source
of discrepancies is the limited nature of the binary
Boolean model itself. In some cases, very steep response
curves for gene expression and protein activities are
observed, corresponding well with the binary nature of
the Boolean model. However, the Boolean formalism
lacks the capacity to describe a continuous reality that
cannot be represented in an on/off manner. For example,
it is impossible to distinguish between absence, low
levels and high levels of glucose, three different condi-
tions that trigger different regulatory responses. Thus, a
discretionary approximation that conveniently explains
the biological context has to be made (e.g., for the
Δsnf1Δsnf4 evaluation, low levels of glucose in a carbon-
limited chemostat were approximated by a situation of
absence of glucose). In other instances, the Boolean
formalism may not be sensitive enough to represent
different levels of expression, such as in the case of
regulation of GAL1 by Mig1, Gal4 and Gal80 [49, 50].
Furthermore, when considering inducible proteins that
are expressed at a basal level, a value of zero may
indicate presence at basal level rather than total absence.
In such instances, defining whether a gene is being
expressed (1) or not (0) is somewhat subjective. A
further limitation of the Boolean formalism, particularly
when focusing on logical steady state analysis with no
distinction between processes of different time scales, is
the difficulty of incorporating negative feedback loops.
As discussed above there are difficulties in representing
the negative feedback loop regulating MTH1 expression,
and it is also impossible to represent Mig1 repression of
the MIG1 gene. Such auto-repression cannot be included
by an AND-relationship, since MIG1 (and Mig1) would
then never be active in a logical steady state.
Errors arising from the discrete evaluation of experimental
gene expression data. Conversion of gene expression
changes into discrete Boolean values is a simplification,
which is presumably prone to errors. Nevertheless, we
used commonly accepted thresholds of fold-change and
significance to decide whether a gene is changing its
expression. We have also checked whether choosing
different a and FCmin greatly impacts model evaluation
results (results not shown), and observed that it does
not. We have identified a number of discrepancies likely
to be due to type-2 errors when assigning experimental
variation (e.g., MIG2 and HXT8 in the Δrgt1 case, and
CAT8 and SIP4 in the Δsnf1Δsnf4 case), which show a
regulatory change in the expected direction although
statistically is not significant. However, one must look
into these errors carefully, especially when the signifi-
cance of the experimental change is calculated based on
a high number of replicates, since this may hint at an
error in the underlying hypothesis instead of a type-2
gene expression error.
Situations where high-throughput data goes against
literature-based knowledge. Large errors found in the
model evaluation (|Yi
mod - Yi
exp| = 2) indicate situations
where model prediction and observed changes of gene
expression have opposite signs. This type of error
represents a situation where the hypothesis underlying
the logical model needs to be reconsidered. We have
found this type of error only 3 times (for GAL4 and
MAL13 in Δmig1, MAL33 in Δsnf1Δsnf4). In the case of
the MAL genes, we have already discussed probable
sources of the wrong predictions, namely the incorrect
assumption about the regulatory mechanisms control-
ling the expression of the MAL loci. For the GAL4 gene,
we have observed that gene expression from the Δmig1
transcriptome study directly contradicts other studies.
Although more careful analysis may be advisable, it is
likely this result arises from the microarray experiment
itself, either due to a problem with the array hybridiza-
tion or with the normalization method used at probe-
sets level.
Conclusion
Overall, the Boolean model showed some potential as a
predictive model. The overall success rate (60% for the
entire model, 71% for the restricted model without genes
of dubious regulation and without considering deletion
of highly pleiotropic GRR1) is promising. The observed
errors are most likely due to a combination of lack of
knowledge on the glucose regulatory network and
simplifications required by the Boolean formalism. In
this regard it should be noted that the deterministic and
binary approximation to reality inherent in the Boolean
formalism demands careful interpretation of model
outputs and limits the overall success rate, which may
be achieved. Even though there is much information on
glucose repression in yeast, it is clear from our analysis
that there are still connections and parts that are missing.
Since the model is set up rigorously based on data from
the literature, these inconsistencies seem to be caused by
a combination of contradictions in reported experimen-
tal results and perhaps due to incorrect insights about
the network topology. Future efforts in modeling of
glucose repression may need to take into consideration
the uncertainties concerning the connectivity of the
regulatory network, as well as network dynamics.
Methods to discern these regulatory uncertainties (for
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 11 of 15
(page number not for citation purposes)
example, through the identification of the most probable
regulatory pathway from a set of different mechanistic
pathway models), moreover have the potential to be
used for reverse engineering of signaling and regulatory
networks. Nevertheless, the model presented here
represents a condensed way of organizing regulatory
information on glucose repression, and strongly facil-
itates integration and evaluation of new hypotheses. It
can also serve as the basis for further efforts in modeling
glucose repression signaling and regulatory pathways
using probabilistic and/or dynamic approaches. The
model presented here is thus an important step towards
a holistic understanding of glucose repression in
Saccharomyces cerevisiae, and the model may further be
used for design of new experiments that can lead to a
better understanding of this complex regulatory system.
Methods
Network reconstruction and logical model representation
Glucose repression signaling and regulatory network was
reconstructed from low-throughput data, namely from
biochemical studies and physiological observations
reported in peer-reviewed, original research publications.
All information found relevant regarding glucose repres-
sion regulatory cascades was collected in a database
specifying the species involved (genes, proteins and
metabolites) and the type of regulation exerted among
them. This information was then converted into a logical
hypergraph, representing all interactions between species
in a logical manner, according to the framework
proposed by Klamt et al. [17]. In our context, a
hypergraph is a generalized unipartite directed graph
representation of an interaction network where each
edge (also called hyperarc or hyperedge) connects a set
of start-nodes (tails) to a set of end-nodes (heads). Here,
we consider graphs with one or more start-nodes, but
with a single end-node. Nodes represent the different
species (genes, proteins or metabolites) and hyperarcs
represent the signal flow between species. A logical
hypergraph is a hypergraph where hyperarcs are repre-
sented by Boolean (or logical) equations (Figure 2),
meaning that the state of an end-node can be determi-
nistically found from the state of start-nodes based on
the defined Boolean function connecting these nodes.
Nodes can assume one of two logical states, on (1) and
off (0); a gene can be expressed (1) or not (0) (or, in a
more specialized case, be upregulated (1) or expressed at
a basal level (0)), a protein can be active (1) or not (0), a
metabolite can be available (1) or not (0). Logical states
represent a discrete approximation of a continuous
reality, for example, a discrete approximation of the
sigmoid curve dictated by the Hill equation used to
describe both gene expression and enzyme activity.
Furthermore,
we
notice
that
the
hypergraph
representation of a Boolean network requires all logical
equations to be written in the so-called disjunctive
normal form, which uses exclusively AND, OR and NOT
operators [17] (Figure 2). All network interactions were
therefore converted into Boolean functions written in
disjunctive normal form, and logical rules were intro-
duced based on literature information. The hereby
reconstructed logical hypergraph can easily and unam-
biguously be converted to its underlying interaction
graph by splitting up hyperarcs with more than one tail.
Our logical hypergraph model represents sensing events
(metabolite – protein interactions), signaling cascades
(protein – protein interactions) and regulatory circuits
(protein – gene interactions) related with glucose
repression in S. cerevisiae. For analyses purposes, we
consider the sensing events (sugar availability) as the
input layer of the system, while the expression levels of
the gene nodes are the outputs used in the model
evaluation. Thus, by properly defining an initial state of
the input layer, we can determine the logical steady state
of all nodes in the system given the defined set of logical
rules underlying the hypergraph. Additionally, given an
input node and an output node, we can also perform a
number of structural analyses on the characteristics of
the pathways connecting them. All our functions are
time-independent, as we are only interested in logical
steady state solutions. Logical steady states analyses
simplify the hypergraph setup, since we do not need to
take into consideration the different timescales of
different processes. Moreover, it allows the model steady
states solutions to be evaluated against data from steady
state chemostat cultivation or from the exponential
phase of a batch cultivation (where balanced growth
resulting in appearance of pseudo-steady-state can be
assumed).
Structural and logical steady state analyses
of the network
We used the MATLAB toolbox CellNetAnalyzer 7.0 [17,
51] to perform structural and functional logical steady
state analyses on the established network. Structural
analyses (number of loops and dependency matrix) were
performed on the underlying interaction graph derived
from the hypergraph. We used CNA capabilities to
analyze the overall number of positive and negative
loops between all input nodes and output nodes. We
also determined the dependency matrix, which sum-
marizes the relationship between all ordered pairs of
species in the network. The dependency matrix is based
solely on the topology of the interaction matrix and does
not incorporate information on Boolean relationships
[17, 51], i.e. while the dependency matrix may tell us
that A is an activator of B, it does not tell us whether
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 12 of 15
(page number not for citation purposes)
species C must be present for the activation to take place.
Each matrix element, Dij, tells us whether the network
contains (1) only activating paths, (2) only inhibiting
paths, or (3) both activating and inhibiting paths,
between species i and j. In addition, it tells us whether
negative feedback loops exist that may attenuate the
predicted (1) activating or (2) down-regulatory effects (if
this is the case, species i is referred to as a non-total
activator or inhibitor). The dependency matrix thus
summarizes information on network topology in a very
condensed way. It was particularly helpful in setting up
the underlying interaction graph, and in identifying parts
of the network that were inconsistent with information
in the literature.
Logical steady state calculations were performed based
on the logical hypergraph representation. Briefly, the
logical steady state is the Boolean state that the system
eventually reaches given a fixed input (see [17] for
detail). We used CNA to determine the logical steady
state of all nodes in the system under all logical
combinations of sugars availability (glucose, galactose,
and/or maltose), and for all single gene deletions and
some double gene deletions. In general, all nodes (except
the input layer) are by default unconstrained. A few
species were given a default value of 1 (genes where no
other regulation is considered and genes expressed at
basal levels). Gene deletions (i.e., knockouts) were
simulated by setting the state of the deleted gene to a
fix value of 0. Finally, specific edges were ignored in
logical steady state analysis if the corresponding regula-
tory interactions were comparably weak.
Model evaluation for knockouts
For some of the knockouts we were able to evaluate
model predictions with available gene expression data
from transcriptome studies. We used the results from the
logical steady state analyses for the corresponding
conditions in order to calculate the changes in gene
expression between the simulated wild-type and the
simulated knockout mutant. For wild-type simulations
we obtained, for each species i, a Boolean state Xi,WT Œ
{0,1}. Similarly, each knockout simulations produces a
logical state Xi,KO Œ {0,1}. The variation between these
two conditions is given by the difference Yi
mod = (Xi,KO -
Xi,WT), with Yi
mod Œ {-1,0,1}. If species i is a gene
(transcript), then Yi
mod can be compared with experi-
mental differential gene expression data, and such
comparison allow us to evaluate the predictive capability
of the model. Thus, we used the available transcriptome
data in order to convert experimental gene expression
changes for each gene i into a discrete number Yi
exp Œ
{-1,0,1}, based on their significance of change (p-value
from a Student's t-test) and fold change (defined as FC =
"average expression of gene i in knockout"/"average
expression of gene i in wild-type" if "average expression
of gene i in knockout" = "average expression of gene i in
wild-type", otherwise FC = -1 × "average expression of
gene i in wild-type"/"average expression of gene i in
knockout"). We established a fold-change threshold
([FCmin| = 1.5) and a Student's t-test p-value cut-off (a =
0.05) for all pair-wise gene expression comparisons
between a deletion mutant and its isogenic reference
strain (i.e., wildtype). All genes with p-value < a and FC
≥FCmin (or FC ≤-FCmin) were assigned with a value of
Yi
exp = 1 (or Yi
exp = -1), and 0 otherwise. Overall, the
model prediction capabilites were evaluated based on
the difference |Yi
mod - Yi
exp|, a value of 0 meaning a
correct prediction, a value of 1 implying a small error,
and a value of 2 indicating a large error (model
prediction and experimental results in opposite
directions).
Evaluation of predictive power
The capabilities of the model to make predictions were
evaluated in terms of the achieved percentage of correct
predictions and by testing the results against the values
expected from a model making random predictions. For
each knockout evaluation, the ratio of correct predictions
was calculated as the ratio of the number of genes where
|Yi
mod - Yi
exp| = 0, divided by the total number of genes
evaluated against experimental gene expression data. In
order to remove uncertainty, we also determined the
percentage of correct predictions excluding dubious
interactions. The model predictions were statistical
evaluated in order to assess the probability of having
correct predictions by chance, a high number indicating
a very bad predictive model. In order to do so, our model
predictions were tested against a random model using
the normal approximation to the binomial distribution.
Specifically, using a one-sided alternative, we tested the
null hypothesis that the proportion of correct predic-
tions by the model, p = X/n (X being the number of
successes and n being the total sample size, i.e. the
number of genes tested), is equal to that expected from
the random model, p0. The statistic used is:
Z
X np
np
p
=
−
−
0
0 1
0
(
)
which is a random variable approximated by the
standard normal distribution [52].
Abbreviations
ODE: ordinary differential equations; CNA: CellNetAna-
lyzer (MATLAB toolbox).
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 13 of 15
(page number not for citation purposes)
Authors' contributions
TSC reconstructed the network, performed all the
analyses, and contributed to the writing of the manu-
script. APO conceived, designed and supervised the
study, was involved in discussing results, and contrib-
uted to the writing of the manuscript. JN designed and
coordinated the study. All authors read and approved the
final manuscript.
Additional material
Additional file 1
Commented list of regulatory species and interactions included in the
Boolean model. Table containing all regulatory species considered, a
description of their function, their mode of regulation and respective
references, and, in some cases, additional notes. This table constitutes
the basis for the Boolean associations used in the Boolean model.
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-3-7-S1.pdf]
Additional file 2
Logical equations included in the hypergraph. Table containing all
logical equations included in the computational evaluation of the
hypergraph. In some cases, the logical equations are accompanied by a
note.
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-3-7-S2.pdf]
Additional file 3
Evaluation of the logical state of the system for all gene deletions and
different carbon sources availability. The Excel file sheet 'KO-WT'
contains the evaluation of the model prediction Yi
mod (Yi
mod = Xi,KO
mod -
Xi,WT
mod) for all single gene deletions and few double deletions under all
combinations of available carbon sources used in this study. The sheet
'WT_copy' contains the state of the system for the wild-type (Xi,WT
mod)
under all combinations of carbon sources. The sheet '2KO_Evalution'
contains the state of the system for all gene deletions (Xi,KO
mod) under
all combinations of carbon sources.
Click here for file
[http://www.biomedcentral.com/content/supplementary/1752-
0509-3-7-S3.xls]
Acknowledgements
We thank Steffen Klamt for help regarding CellNetAnalyzer, and for
providing files that accelerated our computations. We thank Kiran R. Patil
for fruitful discussions throughout the work. APO was funded by
Fundação para a Ciência e Tecnologia from the Portuguese Ministry of
Science and Technology (grant no. SFRH/BD/12435/2003).
References
1.
Blom N, Gammeltoft S and Brunak S: Sequence and structure-
based prediction of eukaryotic protein phosphorylation
sites. J Mol Biol 1999, 294:1351–1362.
2.
Ingrell CR, Miller ML, Jensen ON and Blom N: NetPhosYeast:
prediction of protein phosphorylation sites in yeast. Bioinfor-
matics 2007, 23:895–897.
3.
Johnston M: Feasting, fasting and fermenting. Glucose sensing
in yeast and other cells. Trends Genet 1999, 15:29–33.
4.
Rolland F, Winderickx J and Thevelein JM: Glucose-sensing and -
signalling mechanisms in yeast. FEMS Yeast Res 2002,
2:183–201.
5.
Zaman S, Lippman SI, Zhao X and Broach JR: How Saccharomyces
Responds to Nutrients. Annu Rev Genet 2008.
6.
Santangelo GM: Glucose signaling in Saccharomyces cerevi-
siae. Microbiol Mol Biol Rev 2006, 70:253–282.
7.
Carlson M: Glucose repression in yeast. Curr Opin Microbiol 1999,
2:202–207.
8.
Forsberg H and Ljungdahl PO: Sensors of extracellular nutrients
in Saccharomyces cerevisiae. Curr Genet 2001, 40:91–109.
9.
Klein CJ, Olsson L and Nielsen J: Glucose control in Sacchar-
omyces cerevisiae: the role of Mig1 in metabolic functions.
Microbiology 1998, 144(Pt 1):13–24.
10.
Kitano H, Funahashi A, Matsuoka Y and Oda K: Using process
diagrams for the graphical representation of biological
networks. Nat Biotechnol 2005, 23:961–966.
11.
Oda K, Matsuoka Y, Funahashi A and Kitano H: A comprehensive
pathway map of epidermal growth factor receptor signaling.
Mol Syst Biol 2005, 1:2005.
12.
Oda K and Kitano H: A comprehensive map of the toll-like
receptor signaling network. Mol Syst Biol 2006, 2:2006.0015.
13.
Papin JA and Palsson BO: Topological analysis of mass-balanced
signaling networks: a framework to obtain network proper-
ties including crosstalk. J Theor Biol 2004, 227:283–297.
14.
Shmulevich I, Dougherty ER, Kim S and Zhang W: Probabilistic
Boolean Networks: a rule-based uncertainty model for gene
regulatory networks. Bioinformatics 2002, 18:261–274.
15.
Needham CJ, Bradford JR, Bulpitt AJ and Westhead DR: Inference
in Bayesian networks. Nat Biotechnol 2006, 24:51–53.
16.
Sachs K, Perez O, Pe'er D, Lauffenburger DA and Nolan GP: Causal
protein-signaling networks derived from multiparameter
single-cell data. Science 2005, 308:523–529.
17.
Klamt S, Saez-Rodriguez J, Lindquist JA, Simeoni L and Gilles ED: A
methodology for the structural and functional analysis of
signaling and regulatory networks. BMC Bioinformatics 2006,
7:56.
18.
Chaouiya C: Petri net modelling of biological networks. Brief
Bioinform 2007, 8:210–219.
19.
Sackmann A, Heiner M and Koch I: Application of Petri net
based analysis techniques to signal transduction pathways.
BMC Bioinformatics 2006, 7:482.
20.
Li C, Suzuki S, Ge QW, Nakata M, Matsuno H and Miyano S:
Structural modeling and analysis of signaling pathways
based on Petri nets. J Bioinform Comput Biol 2006, 4:1119–1140.
21.
Kaniak A, Xue Z, Macool D, Kim JH and Johnston M: Regulatory
network connecting two glucose signal transduction path-
ways in Saccharomyces cerevisiae. Eukaryot Cell 2004,
3:221–231.
22.
Johnston M and Kim JH: Glucose as a hormone: receptor-
mediated glucose sensing in the yeast Saccharomyces
cerevisiae. Biochem Soc Trans 2005, 33:247–252.
23.
Kim JH, Brachet V, Moriya H and Johnston M: Integration of
transcriptional and posttranslational regulation in a glucose
signal transduction pathway in Saccharomyces cerevisiae.
Eukaryot Cell 2006, 5:167–173.
24.
Kim JH and Johnston M: Two glucose-sensing pathways
converge on Rgt1 to regulate expression of glucose
transporter genes in Saccharomyces cerevisiae. J Biol Chem
2006, 281:26144–26149.
25.
Li FN and Johnston M: Grr1 of Saccharomyces cerevisiae is
connected to the ubiquitin proteolysis machinery through
Skp1: coupling glucose sensing to gene expression and the
cell cycle. EMBO J 1997, 16:5629–5638.
26.
Moriya H and Johnston M: Glucose sensing and signaling in
Saccharomyces cerevisiae through the Rgt2 glucose sensor
and casein kinase I. Proc Natl Acad Sci USA 2004, 101:1572–1577.
27.
Polish JA, Kim JH and Johnston M: How the Rgt1 transcription
factor of Saccharomyces cerevisiae is regulated by glucose.
Genetics 2005, 169:583–594.
28.
Ozcan S, Leong T and Johnston M: Rgt1p of Saccharomyces
cerevisiae, a key regulator of glucose-induced genes, is both
an activator and a repressor of transcription. Mol Cell Biol
1996, 16:6419–6426.
29.
Ozcan S and Johnston M: Two different repressors collaborate
to restrict expression of the yeast glucose transporter genes
HXT2 and HXT4 to low levels of glucose. Mol Cell Biol 1996,
16:5536–5545.
30.
Ozcan S and Johnston M: Function and regulation of yeast
hexose transporters. Microbiol Mol Biol Rev 1999, 63:554–569.
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 14 of 15
(page number not for citation purposes)
31.
Lutfiyya LL and Johnston M: Two zinc-finger-containing repres-
sors are responsible for glucose repression of SUC2
expression. Mol Cell Biol 1996, 16:4790–4797.
32.
Matern H and Holzer H: Catabolite inactivation of the
galactose uptake system in yeast. J Biol Chem 1977,
252:6399–6402.
33.
Schmidt MC, McCartney RR, Zhang X, Tillman TS, Solimeo H,
Wolfl S, Almonte C and Watkins SC: Std1 and Mth1 proteins
interact with the glucose sensors to control glucose-
regulated gene expression in Saccharomyces cerevisiae.
Mol Cell Biol 1999, 19:4561–4571.
34.
Hu Z, Yue Y, Jiang H, Zhang B, Sherwood PW and Michels CA:
Analysis of the mechanism by which glucose inhibits maltose
induction of MAL gene expression in Saccharomyces.
Genetics 2000, 154:121–132.
35.
Naumov GI, Naumova ES and Michels CA: Genetic variation of
the repeated MAL loci in natural populations of Sacchar-
omyces cerevisiae and Saccharomyces paradoxus. Genetics
1994, 136:803–812.
36.
Lutfiyya LL, Iyer VR, DeRisi J, DeVit MJ, Brown PO and Johnston M:
Characterization of three related glucose repressors and
genes they regulate in Saccharomyces cerevisiae. Genetics
1998, 150:1377–1391.
37.
Klein CJ, Rasmussen JJ, Ronnow B, Olsson L and Nielsen J:
Investigation of the impact of MIG1 and MIG2 on the
physiology of Saccharomyces cerevisiae. J Biotechnol 1999,
68:197–212.
38.
Westergaard SL, Oliveira AP, Bro C, Olsson L and Nielsen J: A
systems biology approach to study glucose repression in the
yeast Saccharomyces cerevisiae. Biotechnol Bioeng 2007,
96:134–145.
39.
Nehlin JO, Carlberg M and Ronne H: Control of yeast GAL genes
by MIG1 repressor: a transcriptional cascade in the glucose
response. EMBO J 1991, 10:3373–3377.
40.
Ostergaard S, Roca C, Ronnow B, Nielsen J and Olsson L:
Physiological studies in aerobic batch cultivations of Sac-
charomyces cerevisiae strains harboring the MEL1 gene.
Biotechnol Bioeng 2000, 68:252–259.
41.
Usaite R, Nielsen J and Olsson L: Physiological characterization
of glucose repression in the strains with SNF1 and SNF4
genes deleted. J Biotechnol 2008, 133:73–81.
42.
Covert MW and Palsson BO: Transcriptional regulation in
constraints-based metabolic models of Escherichia coli. J Biol
Chem 2002, 277:28058–28064.
43.
Akesson M, Forster J and Nielsen J: Integration of gene
expression data into genome-scale metabolic models.
Metab Eng 2004, 6:285–293.
44.
Moreno F and Herrero P: The hexokinase 2-dependent glucose
signal transduction pathway of Saccharomyces cerevisiae.
FEMS Microbiol Rev 2002, 26:83–90.
45.
Hu Z, Nehlin JO, Ronne H and Michels CA: MIG1-dependent and
MIG1-independent glucose regulation of MAL gene expres-
sion in Saccharomyces cerevisiae. Curr Genet 1995, 28:258–266.
46.
Charron MJ, Dubin RA and Michels CA: Structural and functional
analysis of the MAL1 locus of Saccharomyces cerevisiae. Mol
Cell Biol 1986, 6:3891–3899.
47.
Wang X, Bali M, Medintz I and Michels CA: Intracellular maltose
is sufficient to induce MAL gene expression in Saccharo-
myces cerevisiae. Eukaryot Cell 2002, 1:696–703.
48.
Charron MJ, Read E, Haut SR and Michels CA: Molecular
evolution of the telomere-associated MAL loci of Sacchar-
omyces. Genetics 1989, 122:307–316.
49.
Johnston M, Flick JS and Pexton T: Multiple mechanisms provide
rapid and stringent glucose repression of GAL gene
expression in Saccharomyces cerevisiae. Mol Cell Biol 1994,
14:3834–3841.
50.
Lohr D, Venkov P and Zlatanova J: Transcriptional regulation in
the yeast GAL gene family: a complex genetic network.
FASEB J 1995, 9:777–787.
51.
Klamt S, Saez-Rodriguez J and Gilles ED: Structural and
functional analysis of cellular networks with CellNetAnaly-
zer. BMC Syst Biol 2007, 1:2.
52.
Johnson R, Miller I and Freund J: Miller & Freund's Probability &
Statistics for Engineers New Jersey: Prentice-Hall; 2005.
Publish with BioMed Central and every
scientist can read your work free of charge
"BioMed Central will be the most significant development for
disseminating the results of biomedical research in our lifetime."
Sir Paul Nurse, Cancer Research UK
Your research papers will be:
available free of charge to the entire biomedical community
peer reviewed and published immediately upon acceptance
cited in PubMed and archived on PubMed Central
yours — you keep the copyright
Submit your manuscript here:
http://www.biomedcentral.com/info/publishing_adv.asp
BioMedcentral
BMC Systems Biology 2009, 3:7
http://www.biomedcentral.com/1752-0509/3/7
Page 15 of 15
(page number not for citation purposes)
|
19144179
|
Gal3p = ( GAL3 AND ( ( ( galactose_int ) ) ) )
MIG3 = NOT ( ( Rgt1p ) )
GAL4 = NOT ( ( Mig1p ) )
SCF_grr1 = ( GRR1 )
Sip4p = ( SIP4 AND ( ( ( Snf1p ) ) ) )
Gal2p = ( GAL2 )
galactose_int = ( galactose_ext AND ( ( ( Gal2p ) ) ) )
MalRp = ( MALR AND ( ( ( maltose_int ) ) ) )
IDP2 = ( Cat8p )
HXT3 = NOT ( ( Rgt1p AND ( ( ( Mth1p ) ) ) ) )
MEL1 = ( ( Gal4p ) ) OR NOT ( Mig1p OR Gal4p )
JEN1 = ( Cat8p )
Gal1p = ( GAL1 )
Rgt2p = ( glucose_ext AND ( ( ( RGT2 ) ) ) )
GAL3 = NOT ( ( Mig1p ) )
SUC2 = NOT ( ( Mig1p ) OR ( Mig2p ) )
HXT4 = NOT ( ( Mig1p ) OR ( Rgt1p AND ( ( ( Mth1p ) ) ) ) )
Mig2p = ( MIG2 )
MALR = NOT ( ( Mig1p ) )
4ORFs = NOT ( ( RGT1 ) )
HXT2 = NOT ( ( Mig1p ) OR ( Rgt1p ) )
Glc7Reg1 = ( GLC7 AND ( ( ( REG1 AND glucose_ext ) ) ) )
maltose_int = ( maltose_ext AND ( ( ( MalTp ) ) ) )
MTH1 = NOT ( ( Mig1p AND ( ( ( Mig2p ) ) ) ) )
MDH2 = ( Cat8p ) OR ( Sip4p )
GAL5 = ( Gal4p )
GAL10 = ( GAL4 )
MalTp = ( MALT )
Mig3p = ( ( MIG3 ) AND NOT ( Snf1p ) )
Gal4p = ( ( GAL4 ) AND NOT ( Gal80p ) )
SFC1 = ( Cat8p )
GAL7 = ( GAL4 )
GAL1 = ( ( Gal4p ) AND NOT ( Mig1p ) )
FBP1 = ( Sip4p ) OR ( Cat8p )
Gal11p = ( GAL11 )
Cat8p = ( CAT8 AND ( ( ( Snf1p ) ) ) )
Rgt1p = ( RGT1 AND ( ( ( Mth1p OR Std1p ) ) ) )
CAT8 = NOT ( ( Mig1p ) )
Mig1p = ( ( MIG1 ) AND NOT ( Snf1p ) )
Std1p = ( ( ( ( ( STD1 ) AND NOT ( SCF_grr1 ) ) AND NOT ( Rgt2p ) ) AND NOT ( Yck1p ) ) AND NOT ( Snf3p ) )
Snf1p = ( ( SNF1 AND ( ( ( SNF4 ) ) ) ) AND NOT ( Glc7Reg1 ) )
Gal80p = ( ( ( GAL80 ) AND NOT ( Gal3p ) ) AND NOT ( Gal1p ) )
HXT5 = NOT ( ( Rgt1p ) )
HXT8 = NOT ( ( Rgt1p ) )
Yck1p = ( YCK1_2 )
PCK1 = ( Cat8p )
Mth1p = ( ( ( ( ( MTH1 ) AND NOT ( SCF_grr1 ) ) AND NOT ( Snf3p ) ) AND NOT ( Rgt2p ) ) AND NOT ( Yck1p ) )
ACS1 = ( Cat8p )
HXT1 = NOT ( ( Rgt1p AND ( ( ( Mth1p OR Std1p ) ) ) ) )
Snf3p = ( glucose_ext AND ( ( ( SNF3 ) ) ) )
SIP4 = ( Cat8p )
MLS1 = ( Sip4p ) OR ( Cat8p )
ICL1 = ( Cat8p ) OR ( Sip4p )
MALS = ( ( MalRp ) AND NOT ( Mig1p ) )
MIG2 = NOT ( ( Rgt1p ) )
|
The Logic of EGFR/ErbB Signaling: Theoretical Properties
and Analysis of High-Throughput Data
Regina Samaga1, Julio Saez-Rodriguez2,3, Leonidas G. Alexopoulos2,3,4, Peter K. Sorger2,3, Steffen
Klamt1*
1 Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany, 2 Department of Systems Biology, Harvard Medical School, Boston,
Massachusetts, United States of America, 3 Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of
America, 4 Department of Mechanical Engineering, National Technical University of Athens, Athens, Greece
Abstract
The epidermal growth factor receptor (EGFR) signaling pathway is probably the best-studied receptor system in mammalian
cells, and it also has become a popular example for employing mathematical modeling to cellular signaling networks.
Dynamic models have the highest explanatory and predictive potential; however, the lack of kinetic information restricts
current models of EGFR signaling to smaller sub-networks. This work aims to provide a large-scale qualitative model that
comprises the main and also the side routes of EGFR/ErbB signaling and that still enables one to derive important functional
properties and predictions. Using a recently introduced logical modeling framework, we first examined general topological
properties and the qualitative stimulus-response behavior of the network. With species equivalence classes, we introduce a
new technique for logical networks that reveals sets of nodes strongly coupled in their behavior. We also analyzed a model
variant which explicitly accounts for uncertainties regarding the logical combination of signals in the model. The predictive
power of this model is still high, indicating highly redundant sub-structures in the network. Finally, one key advance of this
work is the introduction of new techniques for assessing high-throughput data with logical models (and their underlying
interaction graph). By employing these techniques for phospho-proteomic data from primary hepatocytes and the HepG2
cell line, we demonstrate that our approach enables one to uncover inconsistencies between experimental results and our
current qualitative knowledge and to generate new hypotheses and conclusions. Our results strongly suggest that the Rac/
Cdc42 induced p38 and JNK cascades are independent of PI3K in both primary hepatocytes and HepG2. Furthermore, we
detected that the activation of JNK in response to neuregulin follows a PI3K-dependent signaling pathway.
Citation: Samaga R, Saez-Rodriguez J, Alexopoulos LG, Sorger PK, Klamt S (2009) The Logic of EGFR/ErbB Signaling: Theoretical Properties and Analysis of High-
Throughput Data. PLoS Comput Biol 5(8): e1000438. doi:10.1371/journal.pcbi.1000438
Editor: Anand R. Asthagiri, California Institute of Technology, United States of America
Received January 15, 2009; Accepted June 11, 2009; Published August 7, 2009
Copyright: 2009 Samaga et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: RS and SK are grateful to the German Federal Ministry of Education and Research (funding initiatives ‘‘HepatoSys’’ and ‘‘FORSYS’’), to MaCS (Magdeburg
Centre for Systems Biology) and to the Ministry of Education of Saxony-Anhalt (Research Center ‘‘Dynamic Systems’’) for financial support. J.S.R., L.G.A. and P.K.S.
acknowledge funding by NIH grant P50-GM68762 and by a grant from Pfizer Inc. to P.K.S. and D.A.L. The funders had no role in study design, data collection and
analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: klamt@mpi-magdeburg.mpg.de
Introduction
The
epidermal growth factor receptor (EGFR) signaling
pathway is among the best studied receptor systems in mammalian
cells. Signaling through EGFR (ErbB1) and its family members
ErbB2 (Her2/Neu2) ErbB3 and ErbB4 regulates cellular processes
such as survival, proliferation, differentiation and motility and
ErbB receptors are important targets for new and existing anti-
cancer drugs [1,2].
Mathematical modeling of the EGFR system started more than
25 years ago with efforts to describe binding to and internalization
of the receptor [3] that was followed by a variety of dynamic
models that deal with different aspects of the system (reviewed in
[4,5]). Whereas the first EGFR models focused on the receptor
itself – internalization, ligand binding, and receptor homodimer-
ization [6] – later models included downstream signaling events
(e.g. [7–9]). More recent studies also address homo- and hetero-
dimerization among members of the ErbB receptor family and the
effects on downstream of binding to different ligands (of which 13
are known; e.g. [10–13]). All these models describe aspects of
EGFR/ErbB signaling with a set of stoichiometric reactions and
the dynamics of the involved species is described by a set of
ordinary differential equations (ODEs). In order to simulate the
model, the kinetic constants and initial concentrations of the
model have to be known or, more likely, they must be estimated.
Recently, a large-scale map was constructed by Kitano and
colleagues to capture the current state of knowledge about
interactions in the EGFR system as a stoichiometric network
[14]. This model contains no information on the reaction kinetics
and is thus static and cannot be used to perform dynamic
simulations. Nonetheless, the Kitano map provides a reasonably
comprehensive list of molecules and interactions involved in EGF
signaling and represents an excellent starting point for studying its
global architecture [14–16]. Existing ODE-based models cover
only limited parts of the map, and parametric uncertainty present
even in these smaller models suggests that it is not currently
practical to build an ODE model of the entire pathway having
high explanatory and predictive power. Instead, structural and
qualitative (parameter-free) modeling approaches is the tool of
choice. In fact, many important properties of a system rely solely
PLoS Computational Biology | www.ploscompbiol.org
1
August 2009 | Volume 5 | Issue 8 | e1000438
on the often well-known network structure, including many that
govern dynamic behavior; feedback loops, for example, are
captured in the wiring diagram.
Whereas
structural
(stoichiometric)
analysis
of
metabolic
networks is quite well established [17], relatively few efforts have
been made thus far to study qualitatively the propagation of
information in signaling networks. Efforts to date include statistical
analyses of interaction graphs of large-scale protein-protein
networks (e.g. [18]) and other approaches that rely on graph
theory (e.g. [15,19]). Petri net theory [20,21] and constraint-based
modeling [22] have also been used to unravel structural properties
of signal transduction networks.
Boolean (discrete logic) description of interaction networks has
quite a long tradition in theoretical biology. In the past, it has been
mainly applied to random networks [23] or gene regulatory
networks of moderate size (e.g. [24–27]). However, we have
recently developed a Boolean framework that is specifically
tailored to signaling networks. In contrast to gene regulatory
networks, signaling networks are usually structured into input,
processing and output layers. This approach has recently been
applied successfully to a large-scale model of T cell signaling [28],
and used in concert with high-throughput data to analyze cell-
specific network topologies (Saez-Rodriguez et al, in preparation).
Within this framework, we have set-up a logical model of the
main parts of the stoichiometric model of EGFR signaling [14]
and additionally of signaling through ErbB2, ErbB3 and ErbB4.
As mentioned above, the stoichiometric model of Oda et al [14]
does not allow for dynamic simulations. Also functional issues
related to network structure can be studied only to a minor extent
because the stoichiometric model is limited regarding the analysis
of signal flows relevant in signaling networks. By translating the
stoichiometric (mass-flow based) into a logical (signal-flow based)
representation,
we
obtain
an
executable
model
facilitating
functional predictions about input-output responses of a very
complex signaling cascade. Our model comprises 104 species and
204 interactions and is among the largest of a mammalian
signaling network but we have recently become aware of the
interesting work of Helikar et al [29] who also studied a large-scale
Boolean network containing parts of the EGFR/ErbB induced
signaling pathways. Their work focuses on a statistical analysis of
the possible (non-deterministic) discrete behaviors of their Boolean
model. In contrast, our model provides deterministic and testable
predictions about responses and we have verified many using
functional data. In the process, we have uncovered non-obvious
functional properties of the ErbB signaling pathway that are likely
to be biologically significant.
This paper is organized as follows: the first part describes how
we translated the stoichiometric EGFR/ErbB model of Oda et al
[14] into a logical model via a set of general rules. The second part
presents results from a theoretical analysis of the network
including, for example, a characterization of feedback structure
and identification of network components whose behavior is
strongly coupled. The final section describes application of the
logical model to interpret functional data in which primary human
hepatocytes and hepatocarcinoma cell line HepG2 were exposed
to different ErbB ligands in combination with inhibitors of
intracellular signaling kinases. We show that a Boolean model of
ErbB signaling can generate experimentally verifiable predictions
about input-output behavior in the face of perturbation and that
new hypotheses about biological function can be generated
Results
From a stoichiometric to a logical model for EGFR/ErbB
signaling
Based on a stoichiometric model of EGF receptor signaling [14]
and additional information from the literature, we built a logical
model that describes signaling induced by 13 members of the EGF
ligand family through ErbB1-4, leading to the activation of various
kinases and transcription factors that effect proliferation, growth
and survival (see Figure 1 and Table S1). Ligand binding causes
the formation of eight different ErbB-dimers that autophosphor-
ylate and then provide docking sites for adaptor proteins such as
Gab1, Grb2 and Shc, which transmit signals to the small G
proteins Ras and Rac, leading to the activation of MAPK
cascades. Among these, ERK1/2 is the best studied but our model
also comprises the JNK and p38 cascades. Highly interconnected
with the MAPKs and also downstream of the ErbB receptors is
PI3K/Akt
signaling,
another
major branch
of the
model.
Furthermore, activation of different STATs and the PLCc/PKC
pathway are included.
Our model contains most parts of the stoichiometric model of
Oda et al [14]. However, endocytosis, the G1/S transition of the
cell cycle as well as the crosstalk with the G protein coupled
receptor signaling cascade are not considered in our model as we
focus here on early signaling events induced by external stimuli
(EGF-type ligands). In contrast, our model considers signaling
through
all
different
ErbB
dimers
(in
addition
to
EGFR
homodimers), which was not part of the stoichiometric model
(though a simplified diagram has been given in [14]). Finally, there
are some reactions and species that are only contained in the
logical model so as to use the data set (e.g. the mammalian target
of rapamycin (mTOR), p70S6 kinase). Differences between the
stoichiometric
and
the
logical
model
regarding
considered
components and interactions are also explained in the model
documentation (see Table S1).
Translating a stoichiometric model into a logical model is not a
trivial task and requires additional information. Whenever a species
is only influenced by one upstream molecule, the interpretation as a
Boolean function is straightforward: the downstream species is
Author Summary
The epidermal growth factor receptor (EGFR) signaling
pathway
is
arguably
the
best-characterized
receptor
system in mammalian cells and has become a prime
example for mathematical modeling of cellular signal
transduction. Most of these models are constructed to
describe dynamic and quantitative events but, due to the
lack of precise kinetic information, focus only on certain
regions of the network. Qualitative modeling approaches
relying on the network structure provide a suitable way to
deal with large-scale networks as a whole. Here, we
constructed a comprehensive qualitative model of the
EGFR/ErbB
signaling
pathway
with
more
than
200
interactions reflecting our current state of knowledge. A
theoretical analysis revealed important topological and
functional properties of the network such as qualitative
stimulus-response behavior and redundant sub-structures.
Subsequently, we demonstrate how this qualitative model
can be used to assess high-throughput data leading to
new biological insights: comparing qualitative predictions
(such as expected ‘‘ups’’ and ‘‘downs’’ of activation levels)
of our model with experimental data from primary human
hepatocytes and from the liver cancer cell line HepG2, we
uncovered inconsistencies between measurements and
model structure. These discrepancies lead to modifications
in the EGFR/ErbB signaling network relevant at least for
liver biology.
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
2
August 2009 | Volume 5 | Issue 8 | e1000438
active (state 1) if and only if the state of the upstream species is 1 (vice
versa if the influence is negative) (see Figure 2A). In some other cases
it is clear how to code the dependency in a logical function – for
example, the formation of a complex (e.g. the heterodimerization of
c-Jun and c-Fos to the transcription factor AP-1 (see Figure 2B) or
binding of a ligand to a receptor), where all involved proteins have
to be present to trigger downstream events and are thus connected
with an AND gate. Furthermore, we use an OR gate whenever a
protein can be recruited through different receptors or adapter
proteins (see Figure 2C).
However, in many cases the stoichiometric information is not
sufficient to approximate the activation level of a species as a
logical function of the states of its upstream effectors and one
requires additional (mainly qualitative) information, which can
often be obtained from the literature.
The two main cases that can arise are the following:
N A species is positively influenced by two (or more) upstream
molecules, for example a protein that can be phosphorylated
by different kinases (see Figure 2D). Here, the decision whether
both kinases are necessary or if one suffices, that is whether to
use an AND or an OR, cannot be made on the basis of the
information that is contained in a stoichiometric model.
However, the necessary information can often be obtained
from related literature (e.g. from knock-out studies where one
of both effectors has been removed, or if an inhibitor is
available for an upstream species).
N A species is positively influenced by one species (for example a
kinase) and negatively influenced by another (for example a
phosphatase). In this case, we cannot be sure what happens
Figure 1. Logical model of the EGF-/ErbB receptor signaling pathway represented in ProMoT. Blue circles symbolize AND connections.
Inputs with default value 0 are indicated with red diamonds, inputs with default value 1 by green diamonds. Yellow diamonds stand for the outputs
of the model. Gray hexagons represent the receptors (homodimers as well as heterodimers) and green hexagons stand for the 13 different ligands.
Green ellipses symbolize reservoirs. The remaining species (symbolized with rectangles) are colored according to their function: red: kinases; blue:
phosphatases; yellow: transcription factors; green: adaptor molecules; violet: small G proteins as well as GAPs and GEFs; black: other. The box in the
upper part of the network contains binding of the ligands to the receptor and receptor dimerization, showing the high combinatorial complexity.
Black arrows indicate activations, red blunt-ended lines stand for inhibitions. Dotted lines represent ‘‘late’’ interactions (with attribute t = 2) that are
excluded when studying the initial network response. Dashed lines indicate connections from reservoirs. Dummy species (see Methods) are not
displayed.
doi:10.1371/journal.pcbi.1000438.g001
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
3
August 2009 | Volume 5 | Issue 8 | e1000438
Figure 2. Examples illustrating the translation of the stoichiometric EGFR model into a logical description. The examples are taken
from the stoichiometric map of Oda et al [14]. A The activation level of MKK7 is only influenced by one upstream molecule (active MEKK1). B c-Jun
and c-Fos form the transcription factor AP-1. Accordingly, both species are combined with an AND gate (denoted by ‘‘?’’ in the logical equations). C
Gab1 can bind directly to EGFR homodimers or via receptor-bound Grb2. For the activation of downstream elements, the activation mechanism of
Gab2 does not make a difference what results in a logical OR connection represented by two (independent) activation arrows: Grb2RGab1 OR
EGFRRGab1. D In this example, we cannot immediately decide whether both Raf-1 and MEKK1 are necessary for the activation of MKK1 (in the
model description we use the synonym MEK1) or if the activation of one of these two kinases suffices. Further information is required or an ITT gate
can be used (in model M1 we used an OR based on facts published in the literature).
doi:10.1371/journal.pcbi.1000438.g002
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
4
August 2009 | Volume 5 | Issue 8 | e1000438
when both the kinase and the phosphatase are present; it will
depend on the respective strength (described as kinetic
parameters in a quantitative model) and may differ in different
cell types. However, the activation of phosphatases often
occurs as a temporarily secondary event upon stimulating a
signaling pathway (required for switching off the signal). They
may therefore be neglected when considering the early events,
i.e. the initial response of the network that follows upon
stimulation (see below).
We also have to keep in mind that, in all cases, the logical
description is only a discrete approximation of a quantitative
reaction. In those cases where neither an AND nor an OR is a
good approximation, we can use incomplete truth tables [30]. This
operator, herein after referred to as ‘‘ITT gate’’, returns 1 if and
only if all positive arguments are 1 and all negative arguments are
0, and returns 0 if and only if all positive arguments are 0 and all
negative arguments are 1. In all other cases, no decision can be
made and the response of the molecule remains undefined. Using
ITT gates may limit the determinacy of the model (when
performing stimulus-response simulations it can happen that some
states cannot be determined uniquely), but it allows for a safer
interpretation of the results. To illustrate this concept and to
discuss uncertainties in our reconstructed logical model (in the
following referred to as model M1) we consider a model variant
M2 where the activation mechanisms of 14 proteins are described
with
ITT
gates
reflecting
the
uncertainties
in the
logical
description of M1 (see Table S2). In this way model M2 accounts
explicitly for the uncertainties in the logical concatenation of
different signals, however, it cannot account for uncertainties that
are captured in the wiring diagram itself.
Whenever we refer in the following to ‘‘the logical model’’ we
refer to M1 if not stated otherwise.
Once the network construction has been completed, one may
start to perform discrete simulations. We will not study the
transient behavior of the network; instead we propagate the signals
from the input to the output layer. Mathematically, we compute
the logical steady state that follows from exposing the network to a
certain input stimulus (possibly in combination with network
interventions; see Methods). In this way we can analyze the
qualitative input-output behavior of the network. Feedback loops,
which can be identified in the interaction graph underlying the
logical model, may hamper this kind of analysis of the discrete
behavior of logical networks (especially negative feedback loops
[30]). However, herein we will focus on the initial response of the
network nodes induced by external stimulations or perturbations.
Assuming that the system is in a pseudo-steady state at the
beginning, the initial response of a node is governed by the paths
connecting the inputs with this node whereas feedback loops are
secondary events that can only be activated at a later time point
when each node in the loop has exhibited its initial response.
Although path/cycle length is no precise measure for the velocity
of signal transduction, the comparable average length of input/
output paths (19) and feedback loops (17) supports the assumption
that the initial response of the network nodes is dominated by the
input/output paths whereas feedback loops may overwrite the
initial response of the network nodes only after a certain time
period with significant length (again, feedback loops can causally
not be activated before the initial response occurred). To decouple
the initial response from the activity of the feedback loops, we
proceed as follows: we assign to each reaction a time variable t
determining whether the reaction is active/available during the
initial response (i.e. is an early event; t = 1) or not (late event;
t = 2). In each negative feedback loop we identify the node Z that
has the shortest distance to the input layer. This node Z can be
considered as the initialization point of the feedback loop and we
then assign t = 2 to the ‘‘last’’ interaction of the feedback loop
closing the cycle in node Z (i.e. points into Z). For example, in a
causal chain
InputRARBRC--|DRB we would consider DRB as a late
event. In this way we interrupt the feedback loop and the logical
steady states computed in the network reflect the initial response of
the nodes. Strikingly, it is sufficient to consider only four interactions
as late event to break all feedback loops (see below) in the network.
With this acyclic network a unique logical steady state follows for
any set of input values in model M1. The assignment ‘‘late’’ was not
only reasonable for selected interactions in feedback loops, but also
for three interactions involved in negative feed-forward loops down-
regulating the signaling after a certain time. The time variables for
each reaction can be found in Table S1. Although ‘‘late’’
interactions are neglected when calculating the early signal
propagation, they are nevertheless important to describe structural
properties of the network that can be derived from the interaction
graph representation (see below). It is also important to mention that
the logical steady state computed for a given scenario (see Methods)
does not necessarily reflect the activation pattern in the cell at one
particular point of time. Instead, it reflects for each species the initial
response to the stimulus. The time range in which this initial
response takes place can differ for each molecule – typically, a
species situated in the upper part of the network (e.g. a receptor)
responses faster to the stimulus than a species of the output layer
(e.g. a transcription factor).
We set-up models M1 and M2 with ProMoT [31] and exported
the mathematical description as well as the graphical representa-
tion to the analysis tool CellNetAnalyzer (CNA) [32]. The results
obtained with CNA have been re-imported to and visualized in
ProMoT.
The
logical
model
is
represented
as
logical
interaction
hypergraph (see Methods) and contains 104 nodes and 204
hyperarcs (interactions). Seven interactions are configured as late
events (see Table S1), so their time scale is set to 2. Two
interactions are only considered in the analysis of the interaction
graph but excluded in the logical analysis as they do not change
the logical function of their target node or as the exact mechanism
of the interaction is unknown (see Table S1). 28 nodes are inputs of
the model, i.e. their regulation is not explicitly considered in the
model but can be used to simulate different scenarios. Besides
ligands and receptors, these include for example some phospha-
tases with unknown activation mechanism. For all input nodes, a
default value is given in Table S1 (and is indicated in Figure 1) that
is used for the logical analyses unless otherwise specified.
Topological properties of the interaction graph
A logical model in hypergraph form has a unique underlying
interaction graph (see Methods) capturing merely positive and
negative effects between the elements (instead of deterministic
logic functions). Importantly, the usage of ITT gates in model M2
does not change the underlying interaction graph implying that all
results obtained in this section are valid for both M1 and M2. A
graph-theoretical analysis of the interaction graph enables us to
derive important topological properties of the network, indepen-
dently of the Boolean description. For example, the existence of
feedback loops is necessary for inducing multistationarity (positive
feedback loops) or oscillatory behavior (negative loops) of the
dynamic system [33,34]. In our model, the underlying interaction
graph has 236 feedback loops, thereof 139 negative. Strikingly, all
positive feedback loops are composed of a negative feed-forward
and a negative feedback, except one that describes the reciprocal
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
5
August 2009 | Volume 5 | Issue 8 | e1000438
activation of the adaptor protein Gab1 and PIP3, a lipid of the
membrane layer [35]. All negative feedback loops arise from five
mechanisms: (i) the kinases ERK1/2 and p90RSK downregulate
their own activation by phosphorylation of SOS1, a guanine
nucleotide exchange factor (GEF) for Ras, (ii) the phosphatase
SHP1 binds to the autophosphorylated ErbB1-homodimers and
dephosphorylates them, (iii) Ras positively influences its GTPase
activating protein RasGAP via PI3K, (iv) the ubiquitin ligase c-Cbl
binds to ErbB1, leading to degradation of the receptor in the
lysosome and (v) Ras potentiates the Rab5a-GEF activity of Rin1
and thus increases the formation of endocytic vesicles. Therefore,
removing the species Ras and ErbB1-homodimer breaks all
negative feedback loops. As described above, when considering the
early response in the model the ‘‘last’’ interaction closing a
feedback loop is considered as late event (see Table S1). It turned
out that assigning only four interactions the ‘‘late’’ attribute t = 2
suffices not only to break all negative feedback loops, but also the
positive ones, so that no feedback loop remains in the network
when considering the early events.
In terms of graph theory, a feedback loop is (per definition) a
strongly connected subgraph, i.e. if two species A and B are part of
a directed cycle it always holds that there exists a path from A to B
and from B to A. In our model, all feedback loops build up one
strongly connected component consisting of 34 species, meaning
that all feedbacks are coupled.
Figure 3 shows the participation of the different species in the
feedback loops. Remarkably, the small G protein Ras is included
in 98% of the loops, underlining its central role in the regulation of
this network. Ras is a key regulator of cell fate [36] and a known
oncogene in many human cancers [37]. However, the high
number of feedbacks containing Ras in our model can also reflect
the fact that Ras is one of the best studied proteins and therefore
the feedback mechanisms of Ras are possibly better known than
those of other proteins.
Also noteworthy, RN-tre, a GTPase activating protein (GAP)
for Rab5a, is only involved in positive loops, whereas the guanine
nucleotide exchange factor for Rab5a, Rin1, takes only part in
negative feedbacks.
The large size of the network gives rise to a high number of
possible signaling paths along which one node may affect another
one. There are, for instance, 6786 paths (thereof 52% negative)
leading from the input (ligand) EGF to the transcription factor AP-
Figure 3. Species participation in the feedback loops. The darker a species is colored, the more loops it participates in. Colorless species are
not part of feedback loops. All colored species build up one strongly connected component in the underlying interaction graph.
doi:10.1371/journal.pcbi.1000438.g003
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
6
August 2009 | Volume 5 | Issue 8 | e1000438
1 in the output layer. Considering only the early events, 1684
paths remain being 25% of them negative, where all these negative
paths include the node RasGAP.
The information whether a species acts positively (activating)
or/and negatively (inhibiting) on another species, i.e. whether
there is any positive or/and negative path linking the two species,
can be stored and visualized as dependency matrix [30]. The
dependency matrix for the early events contains ambivalent
dependencies (i.e. a node has positive and negative effects on other
nodes) that mainly rely on the negative influence of RasGAP: as it
inhibits Ras, it gives rise to a number of negative paths connecting
the activated receptors with proteins downstream of Ras – in
addition to the positive paths via SOS1, an activator for Ras. Not
considering RasGAP leads to a matrix where only a few
ambivalent interactions occur (see Figure 4): for example, the
receptor ErbB2 is an ambivalent factor for almost all downstream
elements as it is the preferred heterodimerization partner of the
other receptors and thus prevents signaling through various
different
dimers
(for
example,
ErbB1/ErbB3
formation
is
repressed if ErbB2 is present). When all interactions are active,
Figure 4. Dependency matrix D for the early events (influence of RasGAP not considered). The color of matrix element Di j means the
following: green: species i is an activator of species j (there are only positive paths connecting i with j); red: i is an inhibitor of j (there are only negative
paths connecting i with j); yellow: i is an ambivalent factor for j (there are positive and negative paths connecting i with j); black: i has no influence on
j (there is no path connecting i with j). (See also [32]).
doi:10.1371/journal.pcbi.1000438.g004
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
7
August 2009 | Volume 5 | Issue 8 | e1000438
the dependency matrix contains more ambivalent interactions
than it does when considering only the early events.
Note that, except for ambivalent dependencies, the qualitative
effect (up/down) of perturbations can be unambiguously predicted
from the dependency matrix and we will make use of this
technique when analyzing experimental data (see below).
Theoretical analysis of the logical model
Implementing a Boolean function in each node of the
interaction graph enables us to calculate the qualitative network
response to a certain stimulus or perturbation and to predict the
effects of interventions. Given the binary states for the input
variables and optionally for species that have a fixed value (e.g.
simulating a knock-out or knock-in), one determines the resulting
logical steady state by propagating the signals according to the
logical function of the nodes (see Methods).
Using this technique, we determined the network response in
model M1 upon stimulation with the different ligands, again
focusing on the early events (i.e. the interactions with t = 2 were set
to zero). Due to the fact that the resulting network is acyclic (as
explained above), a unique logical steady state follows for any set
of input values in model M1.
We found that the outputs can be divided into two groups: the
majority of the output elements can be activated by all possible
dimers. However, PKC, STAT1, STAT3 and STAT5 can only be
activated through ErbB1-homodimers (PKC, STAT1, STAT3) or
ErbB1-homodimers and ErbB2/ErbB4-dimers (STAT5). Accord-
ingly, stimulation with neuregulins does not result in activation of
the protein kinase PKC and the transcription factors STAT1 and
STAT3, in contrast to stimulation with the other ligands that
activate all output molecules except the pro-apoptotic effect of
BAD which is repressed. This is due to the fact that the
neuregulins, unlike the other ligands, do not bind to ErbB1 and
thus cannot activate ErbB1-homodimers.
Strikingly, despite of the 14 ITT gates in model M2, the logical
steady state in response to ErbB1-homodimers can still be
determined in model M2 and does not differ from M1. This
observation reflects a high degree of redundancy in at least some
parts of the network. The state of each of the different kinases
phosphorylating p38 or MKK4 is for example only dependent on
the activity of Rac/Cdc42 so that these kinases are always
activated together (see below). Thus, the input–output behavior of
the network can be uniquely predicted for all ligands except
neuregulins. In contrast, model M2 fails to predict the response for
some nodes if other dimers (in absence of the ErbB1-homodimer)
are stimulated. This concerns in particular most of the output
nodes; the states of PKC, STAT1, STAT3 and STAT5 can be
determined (as in model M1, these proteins can only be activated
by ErbB1-homodimers, except STAT5 that is ‘‘on’’ in response to
ErbB2/ErbB4-dimers) whereas the state of the other output nodes
cannot be calculated. The indeterminacy of M2 with respect to
stimulations of dimers others than ErbB1-homodimers can be
explained by the uncertainty (ITT gate) in the activation of Rac/
Cdc42.
When performing simulations with M1, we realized that certain
species in the network show strongly coupled behavior. This
guided us to search systematically for equivalence classes of
network nodes whose activation pattern is completely coupled: for
species A and B being elements of the same equivalence class, it
either holds that their states are always the same (A = 0uB = 0,
A = 1uB = 1;
positive
coupling)
or
always
the
opposite
(A = 0uB = 1, A = 1uB = 0; negative coupling) irrespective of
the chosen inputs. In other words, the state of one species in the
equivalence class determines the states of all other species in this
class. Hence, whenever a species of a particular equivalence class is
active, we can conclude that all other species of the same
equivalence class must have been activated (deactivated in case of
negative coupling), at least transiently.
An algorithm to compute the equivalence classes efficiently is
given in the Methods section. In general, equivalence classes can
be computed for a given scenario (defined by a specific (possibly
empty) set of fixed states, typically from input nodes). For this
given scenario we test systematically for each species whether it is
completely coupled with other nodes or not.
This type of coupling analysis is very similar to enzyme (or
reaction) subsets known from metabolic networks [38,39] and it
helps to uncover functional couplings embedded in the network
structure. We anticipate that the concept of equivalence classes
also provides a basis for model reduction (e.g. when computing
logical steady states), similar as it has been employed in metabolic
networks (see e.g. [40]).
Figure 5 shows the equivalence classes in the EGFR/ErbB
model for early signal propagation where the states (presence) of all
ligands and receptors were left open (the states of the other inputs
were fixed to their default value as given in the model description
(see Table S1)). We found six equivalence classes, the largest
comprising 24 species. The latter includes parts of PI3K signaling
as well as the Rac induced parts of the MAPK cascades reflecting
the strong coupling of these two major pathways in model M1.
In model M2, this equivalence class splits into three smaller ones
because the ITT gates introduce uncertainties that may lead to a
decoupling of the two pathways. The other equivalence classes of
M2 hardly differ from the ones in M1 (see Figure S1) again
indicating that alternative pathways contribute rather to a higher
degree of redundancy than to a higher degree of freedom
regarding the potential input-output behavior.
Another concept relying on the logical description is the
computation of minimal intervention sets (MIS; [30,32]). An MIS
is a set of interventions that induces a certain response, whereas no
subset of the MIS does (i.e. an MIS is support-minimal). One
application of MIS is to determine failure modes in the network
that lead to an activation of elements of the output layer without
any external stimulation of the cell. In the EGFR/ErbB model we
are interested in failures that stimulate proliferation and growth of
the cell when no ligand is present. Regarding the early events,
constitutive activation of Ras, for example, leads to activation of
the transcription factors Elk1, CREB, AP-1 and c-Myc, the p70S6
kinase, the heat shock protein Hsp27 and represses apoptosis –
without any external stimulus. Besides Ras, it is sufficient to
permanently activate one of the species Gab1, Grb2, PI3K, PIP3
or Shc to activate/inhibit these outputs. In model M2, the minimal
intervention sets to provoke the above mentioned response contain
at least two elements, for example the activation of Grb2 and
Vav2.
These findings show that the network has fragile points where a
mutated protein (e.g. one that is constitutively active) may support
uncontrolled growth and proliferation. However, besides ErbB
signaling, various other pathways are important for the regulation
of growth and apoptosis and a failure in one pathway might be
compensated by another, what makes it important to include these
pathways step by step into our model. Additionally, when building
up the model we did not focus on one certain cell type, but
collected species and interactions that have been detected in
different kinds of cells leading to a kind of ‘‘master model’’. A
model that describes only one cell type would probably include less
interactions (Saez-Rodriguez et al, in preparation), so that a
(constitutive) signal has not such a global (network-wide) influence
as in the master model.
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
8
August 2009 | Volume 5 | Issue 8 | e1000438
Analyzing high-throughput experimental data
One of the strengths of our model lies in the broad range of
pathways it covers and in the easy simulation of the network wide
response to different stimulations and interventions. It is therefore
well-suited
to
analyze
high-throughput
data
where
various
readouts are measured in response to several stimuli and to
perturbations all over the network. Here we discuss the analysis of
two datasets collected in primary human hepatocytes and the
hepatocarcinoma cell line HepG2. In the first set of measurements
- a subset of the ‘‘CSR liver compendium’’ (Alexopoulos et al, in
preparation) - primary cells and HepG2 cells were stimulated with
transforming growth factor alpha (TGFa) and additionally treated
with seven different small-molecule drugs, whereof six inhibit the
activation of nodes considered in our model. For the second data
set, HepG2 cells were stimulated with different ligands of the EGF
family and treated with an inhibitor for PI3K. In both cases, the
phosphorylation state of 11 signaling proteins included in the ErbB
model were measured after 0, 30 and 180 minutes (see Methods
for a more detailed description of the experiments). Here, we only
focus on the early response of the network after 30 minutes
because we want to analyze which proteins become activated at
all. We assume that in hepatocytes only ErbB1 and ErbB3 are
expressed as it has been reported for adult rat liver [41]; thus, for
the analysis of the hepatocyte data, the state values of the other
two receptors (ErbB2 and ErbB4) were set to 0 in the model.
As discussed earlier, our modeling framework is based on two
concepts: (i) the Boolean (logical) description discretizing the
kinetic behavior, and (ii) the underlying interaction graph
reflecting the topology of interactions. This gives rise to two
different approaches for the analysis of the data. First, using the
dependency matrix of the interaction graph, we examined whether
the
experimental
results
are
in
accordance
to
the
causal
dependencies in our network. Second, using the logical model,
we predicted the binary network response to the different
experimental stimuli and compared these predictions with a
discretized version of the data.
Interaction graph-based data analysis
In the experiments, the phosphorylation state of the readouts is
measured in response to a particular set of stimuli by adding
Figure 5. Equivalence classes in the EGFR/ErbB model. Each color represents one equivalence class. Species with no color are not part of any
equivalence class. The states for the ligands and the four receptor monomers are left open, all other inputs are fixed to their default value (see Table
S1), which is indicated by the red (0) and green (1) diamonds. Late events are excluded and therefore shown as dotted lines (see also figure 1).
doi:10.1371/journal.pcbi.1000438.g005
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
9
August 2009 | Volume 5 | Issue 8 | e1000438
certain ligands and/or inhibitors and combinations thereof. For
each pair of treatments it can then be checked whether the ratio of
the measured responses is consistent with the causal dependencies
in the network topology (as captured in the dependency matrix;
Figure 4) or not.
By comparing the measured phosphorylation state of a protein p
under treatment A, Xp(A), with the measured value for p under
treatment B, Xp(B), we can characterize the effect of the difference
of both treatments on the activation level of p. We restrict
ourselves here to comparing treatments that differ only in adding
or removing one ligand or inhibitor, although, in principle, all
possible pairwise comparisons of treatments could be considered.
As an example, assume we compare the phosphorylation state
Xp(A) of protein p in response to a stimulation A, where a ligand l
and inhibitor i were added, with the state Xp(B) of p in response to
treatment B, where only the inhibitor i was added. An increase in
the phosphorylation state of protein p in response to the addition of
the ligand (i.e. Xp(A)/Xp(B).1) indicates that there must be at least
one positive path leading from this ligand to the protein and the
respective entry in the dependency matrix (row l, column p) of the
model should therefore show an activating or at least ambivalent
influence.
Analogously, for studying the influence of a certain inhibitor, a
decrease (increase) in the data in response to inhibiting a certain
protein indicates that there must be at least one positive (negative)
path leading from the inhibited species to the respective readout.
We decided to consider a change in the data as significant if
Xp(A)/Xp(B).1.5 or if Xp(A)/Xp(B),1/1.5. Figures 6 and 7 show
the comparison of the data with the dependency matrix of the
model where we considered only the early events and neglected
the influence of RasGAP (as discussed above).
All in all, the experimental network response to the different
treatments agrees reasonably well with the structure of the model,
in particular in primary cells. In HepG2 cells, 10% of the analyzed
dependencies are contradictory to our model: in 3% (7%) of the
cases we saw a significant increase (decrease) in the activation level,
although this was excluded by the model. 45% of the cases agreed
explicitly with the model: in 28% (5%) of the cases, treatments that
have a purely positive (negative) influence according to the
dependency matrix resulted in a significant increase (decrease) in
the measured activation levels and in 12% of the cases a ligand/
inhibitor causes no significant change in a measured readout as
predicted in the model. In the remaining 45% of the cases (gray
entries in Figure 7), the data show no significant change, although
the stimulus can affect the readout in our model (many of these
gray entries will be discussed below). In primary cells, 13% of the
predictions were false, 74% were fully correct and for 13% we
observed no significant changes, although the model contains
paths between the stimulus and the readout. A discussion of
specific findings is given below together with the result of the
logical model.
Data analysis with the logical model
Whereas the dependency analysis described above is based on
the raw data, a comparison of the data with the binary network
response of the logical model requires a discretization of the data,
the simplest being a binarization. To obtain the discretized values,
we used DataRail, a recently introduced MATLAB toolbox that
facilitates the linkage of experimental data to mathematical models
[42]. It provides a variety of methods for data processing,
including algorithms to convert continuous data into binary values
and to create convenient data structures for the analysis in
CellNetAnalyzer. The discretization depends on three thresholds (p1,
p2, p3) which all have to be exceeded in order to discretize the
measured signal to ‘‘on’’ [42]: the first threshold is for the relative
significance (the ratio between the value at time 1 (in our case after
30 minutes) and the value at time 0), the second threshold ensures
the absolute significance (ratio between the signal and the maximum
value for this signal from all measurements) and the third threshold
ascertains that the signal is above experimental noise. The choice
of the thresholds is quite difficult as no reference data exist that
define when a molecule is ‘‘on’’, that is when it is sufficiently
activated to induce its downstream events. Most likely, the
required level of activation differs from protein to protein and
from cell to cell. However, since no information on these
differences is available and to avoid unnecessary degrees of
freedom, we decided to define the same thresholds for all
molecules and both cell types (p1 = 1.5, p2 = 0.15, p3 = 100).
Figure S2 shows the sensitivities of the binarization with respect to
these three parameters.
For each measured scenario we computed the binary network
response of our model and compared it with the discretized data
(Figure 8). We note that the comparison of the measured ‘‘ups and
downs’’ with the dependency matrix (performed in the previous
section) and the comparison of the discretized data with the
predicted logical response are naturally correlated. However, they
do not lead necessarily to exactly the same results. An example:
assume you have an input stimulus (ligand L) which may activate a
target species S via two independent pathways, one of both leading
over an intermediate species A for which we have an inhibitor I. If
we compare the scenario ‘‘stimulation with L and adding inhibitor
I’’ against ‘‘stimulating with L’’ via dependency analysis we would
expect a decrease in the (non-discretized) activation level of S since
the inhibited species A is an activator for S. However, the
phosphorylation state of S might show no significant change in the
dependency analysis (i.e. leads to a ‘‘gray entry’’ as in Figures 6
and 7) due to the alternative pathway not affected by the inhibitor.
In contrast, if the two pathways from L to S are OR-connected in
the logical model, the latter would still predict S to be ‘‘on’’.
Another difference in the data analysis based on dependency
matrix vs. logical model is that the former compares species states
obtained from two different experiments (e.g. experiment with/
without inhibitor) whereas the logical model gives for each
experiment one (independent) prediction for each species.
As in the case of the dependency analysis, the measured data
agree reasonably well with the predictions of the model M1
(HepG2: 77% correct predictions; primary cells: 90% correct
predictions).
In
Figure
S3,
the
comparison
of
model
M2
with the
experimental data is shown. For primary cells, only 7% of the
states cannot be determined due to the ITT gates, for HepG2
21%. 83% of the predictions for primary cells and 59% for HepG2
were correct. In all cases where a state can be predicted by M2 it
naturally coincides with the prediction from M1 since the latter is
only one special case of all possible behaviors in model M2.
In some cases where we used an ITT gate in model M2, the
logical function can be uniquely determined with the experimental
results confirming some of the deterministic logic gates used in
model M1: for example, the transcription factor CREB can be
activated through the MEK-dependent kinase p90RSK AND/OR
through the p38 dependent MK2. As CREB is still activated both
with MEK inhibitor and with p38 inhibitor, this points to an OR-
connection achieving a match between model predictions and data
in this node. In the same way, we can verify an AND connection
for the two negative modulators of Gsk3 and an OR for the
phosphorylation of the auto-inhibitory domain of p70S6 kinase.
Again, using ITT gates, we can only reflect uncertainties
regarding the logical combination of different paths and not
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
10
August 2009 | Volume 5 | Issue 8 | e1000438
whether a species influences another at all. This is why some of the
discrepancies between the predictions of model M1 and the data
also appear for model M2.
Interpreting inconsistencies between data and model
predictions
Most disagreements between model predictions and experi-
mental results concentrate on certain experimental conditions
(rows) and readouts (columns) - in the dependency analysis as well
as in the analysis with the logical model. Here we discuss such
systematic inconsistencies and – using our model – we seek to
provide explanations and conclusions:
N A significantly increased state of phosphorylation of STAT3 in
response to any of the ligands could not be found both in
HepG2 and primary hepatocytes. Whether this is due to the
fact that the activation of STAT3 is very transient, as it has
been reported for example for the human epithelial carcinoma
cell line A431 [43], or if the activation of this transcription
factor through ErbB receptors plays no role in hepatocytes, has
still to be clarified.
N Both analysis approaches show that stimulation of HepG2 cells
with amphiregulin (not measured in primary cells) did not
result in activation of the measured proteins (see Figure 7, lines
34–37 and Figure 8B, lines 23/24). This is in agreement with
Figure 6. Interaction graph-based comparison between experimental data and topological properties of the model (data from
primary hepatocytes). Shown is the comparison between the measured and predicted changes (‘‘ups’’ and ‘‘downs’’) in the activation levels of
network elements in response to ligands and inhibitors in primary human hepatocytes (data obtained from Alexopoulos et al, in preparation). Each
row compares two different scenarios A and B. A dot behind the species name in the row labels indicates that, in both scenario A and scenario B, this
species was added as ligand (green dot) or an inhibitor for this species was added (red dot). Species whose input values differ in both scenarios are
marked with an up or down arrow, respectively. For example, the comparison of scenario A (EGF ligand, TGFa ligand, PI3K inhibitor) and scenario B
(TGFa ligand, PI3K inhibitor) is labeled by TGFa N (green dot), PI3K N (red dot), EGF q, i.e. the influence of an increased level of EGF on the readouts is
analyzed (under the side constraints that TGFa and a PI3K inhibitor were added as well; for further explanations see text). The readouts are shown in
the columns. The color indicates whether the model predictions and the measurements are consistent or not (see color legend).
doi:10.1371/journal.pcbi.1000438.g006
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
11
August 2009 | Volume 5 | Issue 8 | e1000438
Figure 7. Interaction graph-based comparison between experimental data and topological properties of the model (data from
HepG2 cells). Shown is the comparison between the measured and predicted changes (‘‘ups’’ and ‘‘downs’’) in the activation levels of network
elements in response to ligands and inhibitors in HepG2 cells. The horizontal line separates the first (top) from the second (bottom) dataset for
HepG2 cells (see also text). For further explanations and color legend see Figure 6.
doi:10.1371/journal.pcbi.1000438.g007
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
12
August 2009 | Volume 5 | Issue 8 | e1000438
Figure 8. Comparison of the discretized data with predictions from the logical model. A Primary human hepatocytes (data from
Alexopoulos et al, in preparation). B HepG2 cells (the horizontal line separates the first (top) from the second (bottom) dataset for HepG2 cells; see
also text). Each row represents one treatment and the readouts are shown in the columns. Light green: predicted correctly, ‘‘on’’; dark green:
predicted correctly, ‘‘off’’; light red: predicted ‘‘on’’, measured ‘‘off’’; dark red: predicted ‘‘off’’, measured ‘‘on’’, black: data points where the measured
species is inhibited are not considered.
doi:10.1371/journal.pcbi.1000438.g008
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
13
August 2009 | Volume 5 | Issue 8 | e1000438
findings of amphiregulin being a much weaker growth
stimulator than EGF in some cell types [44].
N The systematic errors in the column of p38 in the dependency
analysis (for primary as well as HepG2 cells) might indicate
missing edges in the model requiring further experimental
studies to verify these findings. We cannot exclude that other
(e.g. stress-induced) pathways not captured in our model may
have caused these observations, also because some of the
effects on p38 are also present without ligand stimulation.
N Stimulating the HepG2 cells with both TGFa and EGF does
not result in a significantly higher activation level of the
readouts compared to adding only one of these ligands as can
be seen from the predominantly gray entries in lines 26/27 and
44/45 in Figure 7. This finding is in accordance with the fact
that both ligands are very similar and bind to the same
receptor dimers (see Table S1).
N One of the major differences in the behavior of the two cell
types is the activation of Hsp27: whereas this heat shock
protein becomes activated in response to cytokine stimulation
in primary cells, no significant increase in the state of
phosphorylation occurs in almost all studied scenarios in the
cancer cell line (leading to many false ‘‘on’’ predictions).
N Another remarkable discrepancy between the experimental
data and our model predictions is the influence of the mTOR
inhibitor rapamycin on phosphorylation of p70S6 kinase (see
lines 14/15 in Figures 6 and 7), which is not supported by our
model. Although mTOR mediates the phosphorylation of the
catalytic site T389 [45], it has to the best of our knowledge not
been implicated with the phosphorylation of T421 and S424,
those sites, whose state of phosphorylation were measured in
the analyzed data sets. However, an inhibitory effect of
rapamycin on these sites has been reported earlier [46], even if
the molecular mechanism that could explain this influence still
has to be uncovered.
N According to our model, PI3K should influence all measured
readouts except STAT3. However, the data show a clear effect
of the PI3K inhibitor only on the phosphorylation of Akt (see
Figure 6, lines 12/13 and Figure 7, lines 50–61). Additionally,
Figure 8 shows that JNK, p38 and, in primary cells also
Hsp27, could be activated in the experiments in presence of
PI3K inhibitor although our model predicted the phosphor-
ylation to be blocked (due to the AND connections of the
PI3K-dependent nodes PIP3 and PI(3,4)P2, respectively, with
Vav2 and SOS1_Eps8_E3b1). We therefore searched for
hypothetical changes in our model structure that could explain
these experimental findings. We observed that node Rac/
Cdc42 lies on all paths connecting the inputs (ligands) with the
aforementioned critical readouts (except Gsk3, see below), i.e.
activation of Rac/Cdc42 is necessary in our model for
phosphorylation of JNK, Hsp27 and p38. We may thus
hypothesize that - in contrast to the assumption in our model -
PI3K activity is not necessary for activation of the small G-
proteins Rac and Cdc42 in primary hepatocytes and in
HepG2 cells.
N A closer look on Figure 8B (lines 19/20) reveals that the
phosphorylation of JNK in response to neuregulin is – in
contrast to the response to any of the other ligands – sensitive
on PI3K inhibitor. This is also reflected in Figure 7 where an
increase of neuregulin only increases the phosphorylation of
JNK in absence of PI3K inhibitor (see lines 28–33) and
decreasing the level of PI3K (i.e. adding the inhibitor) after
neuregulin stimulation also leads to a decreased phosphoryla-
tion state of JNK (see lines 52 and 59). Therefore, neuregulin
must use a different, PI3K dependent signaling path for
activating JNK than the other ligands, probably due to the fact
that neuregulin only activates ErbB1/ErbB3-dimers whereas
EGF, TGFa, amphiregulin and epiregulin additionally
activate ErbB1-homodimers. Taking these findings together,
we propose the following alternative mechanism: Vav2 is the
major GEF for Rac/Cdc42 in hepatocytes and activates Rac/
Cdc42 in a PI3K-independent way. Neuregulin, which cannot
bind to ErbB1-homodimers and accordingly is not able to
activate Vav2 (see Table S1), provokes the activation of JNK
independently of the Rac/Cdc42 induced MAPK cascade
through a different, PI3K-dependent pathway.
N In the model, the inhibitory phosphorylation of Gsk3 can be
induced by a MEK1/2 dependent pathway (via p90RSK) and
by a PI3K dependent pathway (via Akt). Figures 6 and 7 (lines
9 and 13) show that the phosphorylation of Gsk3 in response to
TGFa is independent of the MEK inhibitor and the PI3K
inhibitor, both in HepG2 and in primary cells. As TGFa
stimulation leads to a strong phosphorylation of Gsk3 in both
cell types (see Figure 8), there must be another signaling route,
not involving MEK and PI3K. One possible candidate is PKC
which has already been reported to inhibit Gsk3, however not
in response to ligands of the EGF family [47].
N According to the data, both Gsk3 and p90RSK are influenced
by JNK inhibitor after TGFa stimulation in primary
hepatocytes (see Figure 6, line 18). This seems to support
another possible mechanism, where JNK activates p90RSK
which may then phosphorylate Gsk3. However, the JNK
inhibitor affects much more proteins than expected, both in
HepG2 and in primary cells. As these unexpected influences
also occur in absence of ligand stimulation, this strongly
suggests a minor specificity of the JNK inhibitor.
N Similar as for Gsk3 phosphorylation, data analysis with our
model provides useful insights into the activation mechanism
of CREB in response to TGFa: the proposed effect of the p38
dependent kinase MK2 on CREB cannot be observed both in
HepG2 and in primary cells (see Figures 6 and 7, line 11). The
positive effect of MEK on CREB phosphorylation after TGFa
stimulation can be seen in HepG2 (Figure 7, line 9), but not in
primary hepatocytes (Figure 6, line 9). Together with the
finding of the logical analysis that the MEK inhibitor cannot
block activation of CREB in HepG2 (Figure 8), this indicates
that there must be an alternative pathway for CREB activation
in primary hepatocytes that is probably involving p90RSK.
A summary of the above mentioned results is given in Table S3.
Changing the model accordingly, we can improve the agreement
of model predictions and data in the logical analysis from 90% to
97% for the primary cells and from 74% to 94% for HepG2. For
the dependency analysis, the number of comparisons that agree
explicitly increases from 74% to 82% for primary and from 45%
to 64% for HepG2 cells. Moreover, the number of entries where
we assumed a change in the data but could not detect a significant
increase or decrease reduces from 13% to 4% (primary) and from
45% to 24% (HepG2), albeit at the expense of a minor increase in
the number of contradictions (primary: increase from 13% to
14%, HepG2: 10% to 12%).
As described above, herein we deduced the proposed changes of
the model structure manually from the data analysis. More
systematic approaches for network identification from combina-
torial
experiments
are
given
in
Saez-Rodriguez
et
al
(in
preparation) and in [48].
In general, detecting such systematic inconsistencies of the data
both with respect to the dependency structure of the network and
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
14
August 2009 | Volume 5 | Issue 8 | e1000438
the logical model description is a great advantage of our approach
and could hardly be achieved with a model relying on differential
equations (where parameter uncertainty often hampers a falsifica-
tion of the model structure).
Discussion
In the present work, we developed a large-scale logical model of
signaling through the four ErbB receptors, including the ERK,
JNK and p38 MAPK cascades, Akt signaling, activation of STATs
and the PLCc pathway, based on the stoichiometric pathway map
of Oda et al [14]. We discussed technical problems that arise when
converting a stoichiometric model into a logical one and proposed
a general guideline how to deal with them.
We examined several properties of the logical model charac-
terizing its topology (feedback loops and network-wide interde-
pendencies as derived from the underlying interaction graph) and
its qualitative input-output behavior with respect to different
stimuli. We also introduced the new technique of species
equivalence classes revealing coupled activation patterns in the
logical model providing valuable insights into the correlated
behavior of network elements.
One possibility to deal with uncertainties concerning the correct
logical combination of different influences on a certain node is the
usage of gates with incomplete truth tables (ITT gates). We
replaced the (deterministic) logical gates for the activation of 14
species of our model with ITT gates and repeated all logical
analyses with this modified model. Surprisingly, the predictive
power of the ITT model is still high, highlighting the redundant
structure of major parts of the signaling pathway and showing that
many properties of the network do not rely on the assumptions we
made when choosing the logical functions.
Compared with a dynamic model based on differential
equations, our approach for describing signaling events is certainly
limited in reflecting kinetic aspects which are important to obtain a
complete understanding of these processes in the cell. However,
properties derived exclusively from the structure can provide
insights into the transfer of signals in the cell, as the result of this
and other studies have shown [28,29]. The simpler design of the
qualitative models also has some advantages over complex
dynamic models. First of all, the logical approach enables us to
model large-scale signaling networks allowing, for example, to
study the effects of crosstalk, for which a dynamic description is
currently often unimaginable. An expansion of the model can
easily be done, whereas adding a reaction to a model of differential
equations requires usually the elaborate re-estimation of param-
eters. The flexible architecture of the model also enables us to test
and generate hypotheses very quickly. Another advantage is that
the qualitative predictions derived with a logical model do not
depend on certain parameter values except the time scales and are
therefore more generally valid. There are also methods to study
ODE models without parameters (e.g. [49–51]). However, these
methods are currently limited to relatively small systems and study
different properties.
With the advances of experimental techniques, it becomes more
and more essential to provide tools that allow for the analysis and
exemplification of the huge amount of data that arise. We
developed new techniques for the analysis of large data sets that
are especially
well-suited to
analyze data that
stem from
combinatorial experiments (systematic combination of different
ligands/inhibitors). The first approach, a method for comparing
experimental (high-throughput) data with predictions derived from
the logical model, requires a discretization of the data. Although
the ‘‘on/off’’ decision is sometimes hard to take as no reference
data exist and the ‘‘right’’ thresholds for the parameters are
unknown, assessing the sensitivities of the data with respect to the
discretization thresholds leads to a safer interpretation. Alterna-
tively, the data can be assigned a relative value between 0 and 1
which can be compared to the discrete (0/1) value of the model
(Saez-Rodriguez et al, in preparation). The second approach, the
comparison of the data with the topological dependency structure
of the model (captured in the interaction graph), requires only a
significance threshold and provides an even simpler method for
the falsification of qualitative knowledge as it relies on less
assumptions than the logical model (only the wiring diagram is
evaluated; logical combinations and discrete states are not
required).
Applying these new automatized techniques to analyze high-
throughput phospho-proteomic data revealed some important
insights into the structure of EGFR/ErbB signaling in primary
hepatocytes and the HepG2 cell line. Our results strongly suggest a
model where the Rac/Cdc42 induced p38 and JNK cascades are
independent of PI3K, both in primary hepatocytes and in HepG2.
Furthermore, we detected that the activation of JNK in response
to neuregulin follows a PI3K-dependent signaling pathway that
seems not to be important for activation of JNK through ErbB1-
binding ligands. Additional findings concern Gsk3 and CREB
where known signaling paths were excluded to provoke phos-
phorylation after TGFa stimulation and new routes could be
proposed. Finally, we observed no activation of STAT3 in both
cell types and no activation of Hsp27 in HepG2. Besides these
results on the topology of EGFR/ErbB signaling in hepatocytes,
the comparison of model predictions and data could also detect
side effects of the used JNK inhibitor.
With our software CellNetAnalyzer (CNA; [32]) we provide a
powerful tool to study structural networks. It facilitates the analysis
of interaction graphs as well as logical models and also provides
methods to compare model predictions with experimental data as
described herein. Furthermore, CNA is now highly coupled with
the tools ProMoT [31], DataRail [42] and CellNetOptimizer (Saez-
Rodriguez et al, in preparation), forming an integrated pipeline for
the construction, structural analysis and data interpretation of
signal transduction networks.
The presented model is to the best of our knowledge one of the
largest existing mathematical models of the EGFR/ErbB signaling
pathway. However, it is far from being complete and has to be
complemented, for example by including the endocytosis of the
receptors. Step by step, we want to expand the model by other
important mitogenic and pro- and anti-apoptotic pathways to
study crosstalk. We also think that the logical model can serve as a
useful basis for the development of dynamic models. A step
between both modeling frameworks could be to refine the current
binary description and use multilevel activation instead, a
promising approach yet it requires more detailed (semi-quantita-
tive) information on the reaction kinetics and leads to more
complex networks. Further refinements could be achieved by fuzzy
logic description or by considering more precise time delays for the
interactions.
Methods
Logical modeling of the EGFR/ErbB signaling network
For the reconstruction and qualitative analysis of the EGFR/
ErbB signaling network we employ a logical modeling framework
as introduced previously [30,32]. Signaling networks are usually
structured into input, intermediate and output layer and the input
signals govern the response of the network. For this characteristic
network topology we introduced logical interaction hypergraphs (LIHs)
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
15
August 2009 | Volume 5 | Issue 8 | e1000438
as a special representation of Boolean networks, which is well-
suited to formalize, visualize and analyze logical models of signal
transduction networks. As in all Boolean networks, nodes in the
network represent species (e.g. kinases, adaptor molecules or
transcription factors) each having an associated logical state (in the
binary case as used herein only ‘‘on’’ (1) or ‘‘off’’ (0)) determining
whether the species is active (or present) or not. Signaling events
are encoded as Boolean operations on the network nodes. For
example, the MAP kinase (MAPK) JNK can be activated (gets
‘‘on’’) if the MAPK kinase MKK7 AND the MAPK kinase MKK4
are active (see the AND connection in Figure 1). Usually, a node
can be activated by more than one signaling event; all these events
are then OR-connected, e.g. the MAPK p38 becomes active if
MKK3 OR MKK4 OR MKK6 is active (Figure 1).
In general, in LIHs we make only use of the Boolean operators
AND (?), OR (+), and NOT (!), which are sufficient to represent
any logical relationship. A signaling event (or interaction) in an
LIH is an AND connection of nodes (negation of node values using
the NOT operator are allowed) describing one opportunity how
the target species of this connection can be activated. Hence, for
the first example described above we would write
MKK7 AND MKK4?JNK
or shorter
MKK7:MKK4?JNK
In a graphical representation of the network (see JNK node in
Figure 1), such an AND connection is displayed as a hyperarc. In
contrast to arcs in graphs, a hyperarc (in hypergraphs) may have
several start or end nodes. Clearly, in some cases, only one species
is required to activate another, as in the example
MKK3?p38:
In these cases, the hyperarc is a simple arc as occurring in graphs; we
will nevertheless refer to it as a hyperarc. As already mentioned, a
species may be activated via several distinct signaling events
(hyperarcs), i.e. all these signaling events are OR-connected. This
can again be illustrated by p38, which can be activated (indepen-
dently) via three different MAPKs and we therefore have three
different OR-connected hyperarcs:
MKK3?p38
OR
MKK4?p38
OR
MKK6?p38
Hence, all hyperarcs pointing into a species are OR connected. In this
way we can easily interpret Figure 1, which displays graphically the
interactions given in Table S1.
As described in the main part, the reconstruction of our logical
model of EGFR/ErbB is based on a stoichiometric model of EGF
receptor signaling [14] and additional information from the
literature. Some general remarks on how a stoichiometric network
can be translated into a logical one are given in the main part. The
logical model (for both version M1 and version M2; the latter
having 14 gates with incomplete truth tables; see main text)
comprises signaling of 13 members of the EGF ligand family
through the EGF receptor and its heterodimerization partners
ErbB2-4, leading to the activation of various transcription factors
and kinases that effect proliferation, growth and survival (Figure 1).
In addition to ligands and receptors, species whose regulation is
not known are herein considered as members of the input layer,
for example the phosphatases PTEN and SHIP2.
The differentiation between ‘‘early’’ and ‘‘late’’ events (see
below and main part) makes it sometimes necessary to introduce
auxiliary (‘‘dummy’’) nodes that have no biological correspon-
dents. Consider for example a species C that is activated by species
A during the early events (t = 1) and down-regulated by another
species B as a late event (t = 2). Assuming that both the presence of
A and the absence of B are necessary to activate C, we use an
AND connection in the LIH representation (A ? !BRC). As the
two influences are combined to one hyperarc in the LIH, we can
assign only one time variable to this interaction. In order to reflect
the time delay of the inhibitory activity of B, we introduce an
additional dummy node with t = 2. We now describe the original
interaction A ? !BRC with two interactions
B?B dummy t~2
ð
Þ
A:!B dummy?C t~1
ð
Þ:
An example in the ErbB model are the ErbB1-homodimers that
are activated by various ligands (e.g. EGF) and dephosphorylated
by SHP1 (see Table S1). To properly describe the timing of the
SHP1-mediated dephosphorylation of the receptor, we introduce a
dummy species shp1d that is activated by SHP1 and obtain thus
two hyperarcs:
shp1?shp1d t~2
ð
Þ
egf:erbb1:!shp1d?erbb11 t~1
ð
Þ:
Another type of node that is introduced for modeling purpose only
is what we refer to as reservoir. It is used whenever a molecule
causes different downstream events depending on how it is
activated. Here, we have to use more than one compound to
describe the molecule in the model. An example in our model is
mTOR: associated with Rictor, it is involved in the activation of
Akt, whereas the Raptor-bound form activates p70S6 kinase.
However, as all these compounds represent the same biological
species, we associate them with a reservoir, pointing out that they
share the same pool. Inactivation of the reservoir will then affect
the activation of all correspondents of this species.
A full description of the model M1 with all species and
interactions (hyperarcs) is given in Table S1. In model variant M2,
14 logical gates of model M1 have been configured as incomplete
truth tables (ITT gates). The differences between M1 and M2 are
described in Table S2.
Analysis of the logical model
Once an LIH has been set-up, we may start to analyze it. A
typical scenario is that we apply a pattern of inputs to the network
and we would like to know how the nodes in the network will
respond to this stimulation. As explained in [32], by propagating
input signals along the logical (hyperarc) connections (which is
equivalent to computing the logical steady state resulting from the
input stimuli) we obtain the qualitative response of the network.
Note that the logical steady state obtained by this propagation
technique is independent of the assumption of synchronous or
asynchronous switching which is required when analyzing the
discrete dynamics of Boolean networks [27]. It depends on the
functionality of positive or negative feedback loops in the network
whether we can resolve a complete and unique logical response of
all nodes for a given set of input stimuli (for example, negative
feedback loops may prevent the existence of a logical steady state).
Feedback loops are usually present in signaling networks, however,
as described in the main part, we identified one interaction in each
loop that can be considered as a late event (t = 2). When
considering the initial response of the network we set these late-
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
16
August 2009 | Volume 5 | Issue 8 | e1000438
event connections inactive leading to an acyclic network for which
always a unique network response for a given set of inputs can be
computed.
One can also easily perform in silico experiments, for example
check how a knock-out (or inhibition) alters the network response
by fixing the state of the respective species.
With the idea of minimal intervention sets (MIS) one may even
directly search for those interventions that enforce a desired
response (e.g. activation or inactivation of a transcription factor).
As described in [32], MISs can be computed by testing
systematically which combinations of knockouts and knockins
fulfill a specified intervention goal.
Species equivalence classes in logical networks
A new analysis technique for logical networks is introduced in this
work: we search for equivalence classes of network nodes whose
activation pattern is completely coupled in logical steady state:
species A and B are elements of the same equivalence class, if it
either holds that their values in steady state are always the same
(A = 0uB = 0, A = 1uB = 1; positive coupling) or always the
opposite (A = 0uB = 1, A = 1uB = 0; negative coupling) irrespec-
tive of the chosen inputs (e.g. ligands). In other words, the state of
one species in the equivalence class determines the states of all other
species in this class. Again, the relation given above holds for logical
steady states where both A and B are determined and where no
intervention was made in the network except for the inputs.
Whenever a species of a particular equivalence class is active, we
can conclude that all other species of the same equivalence class
must have been activated (deactivated in case of negative
coupling), at least transiently.
An efficient algorithm for computing the equivalence classes can
be constructed as follows:
1)
Equivalence classes can be computed for a given scenario, so
we first define a specific (possibly empty) set of fixed states,
typically from (some) input nodes.
2)
For this given scenario we test systematically for each species
whether it is strongly coupled with other nodes or not,
independently of external stimuli. For each species A we
compute (i) the logical steady states of all other species that
result when fixing the state of A to 1 and (ii) the logical steady
states of all other species that result when fixing the state of A
to 0. A node B whose logical steady state can be determined
in both cases and is 1 in one case and 0 in the other case is
known to be in one equivalence class with species A: B is
positively coupled with A if the two resulting logical steady
states of B are 1/0 (it then holds A = 1 = .B = 1,
A = 0 = .B = 0 and thus according to contraposition also
B = 0 = .A = 0, B = 1 = .A = 1) and negatively coupled if the
two logical steady states are 0/1 (it then holds A = 1 = .B = 0,
A = 1 = .B = 0 and thus according to contraposition also
B = 0 = .A = 1, B = 1 = .A = 0). The case that the logical
steady state of a species B is 0/0 or 1/1 (for fixing A = 1/
A = 0) indicates that this species B can never be activated or
never be inhibited, respectively, and would thus indicate a
semantic problem in the model.
If a species A is coupled with species B, and species B is coupled
with species C, we can subsume all three species in one equivalence
class (we do that systematically for all species until we reach finally
the equivalence classes). Composing the equivalence classes in this
way, it may also happen that species that cannot influence each
other (no directed path between both exists) are in one equivalence
class due to a common upstream regulator. Consider a network
that only contains the interactions A R B and A R C. Fixing the
state of B or C to 1/0 we cannot conclude any equivalence
relations as no further states can be determined. Fixing A to 1
and 0 we find that A is equivalent to B and A is equivalent to C,
thus – according to the rule given above – A, B and C form one
equivalence class.
Interaction graph analysis
Another advantage of LIHs is that we can easily derive the
(signed and directed) interaction graph underlying the logical
model: we only have to split all hyperarcs that have two or more
start nodes (i.e. the AND connections) into simple arcs. Interaction
graphs cannot be used to give on/off predictions; however, they
provide an appropriate formalism to search for signaling paths and
feedback loops. Another useful feature that can be extracted from
interaction graphs is the dependency matrix as introduced in [30,32]
which displays network-wide interdependencies between all pairs
of species. For example, a species A is an activator (inhibitor) of
another species B, if at least one path leads from A to B and if all
those paths are positive (negative). This kind of information can be
very useful for predicting effects of perturbations.
Model implementation and availability
We set-up the logical EGFR/ErbB model with ProMoT [31] and
exported the mathematical description as well as the graphical
representation to the analysis tool CellNetAnalyzer (CNA) [32]. The
results obtained with CellNetAnalyzer have been partially re-
imported to and visualized in ProMoT (Figures 1, 3, 5). Data
management and discretization was performed with DataRail
[42].
The tools are freely available (for academic use) from the
following web-sites:
DataRail: http://code.google.com/p/sbpipeline/wiki/DataRail
ProMoT: http://www.mpi-magdeburg.mpg.de/projects/promot/
CellNetAnalyzer: http://www.mpi-magdeburg.mpg.de/projects/
cna/cna.html
After acceptance, the model will be provided in formats for
ProMoT and CellNetAnalyzer.
Experimental set-up and measurement data
The data on primary human hepatocytes and the first part of
the HepG2 data were obtained from experiments conducted by
Alexopoulos et al (in preparation), while for the second part of the
HepG2 data, a cue-signal-response (CSR) compendium was
created for the EGFR pathway. The second dataset comprises
11 phosphoprotein measurements under 24 different perturbations
generated by the combinatorial co-treatments with a diverse set of
ErbB ligands and the PI3K inhibitor. For ligands we choose 5
ErbB related cytokines, namely epidermal growth factor (EGF),
neuregulin 1 (NRG1; also known as heregulin), amphiregulin
(AR), epiregulin (EPR), and transforming growth factor alpha
(TGFa). For each stimulus, the PI3K inhibitor ZSTK-474 was
added at 2 mM final concentration 30 minutes prior to any ligand
treatment. Optimal inhibitor concentration was obtained for
concentration-inhibition curve (data not shown) in order to
achieve 95% inhibition of the downstream pAkt signal on TGFa
stimulated HepG2. The dataset was created using a high-
throughput method of bead-based fluorescent readings (Luminex,
Austin, TX). Assays were optimized for multiplexability and
checked for passage-to-passage and preparation-to-preparation
variability (Alexopoulos et al, in preparation).
The full dataset (first and second part) and the resulting
discretization are graphically depicted in Figure S4.
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
17
August 2009 | Volume 5 | Issue 8 | e1000438
Supporting Information
Figure S1.
Equivalence classes for model M2. Each color
represents one equivalence class. The equivalence classes of model
M1 are depicted by the species border color. Late interactions
(t = 2) are drawn as dotted lines. The value of fixed inputs is given
by the green (1) and red (0) diamonds.
Found at: doi:10.1371/journal.pcbi.1000438.s001 (0.33 MB PDF)
Figure S2.
Sensitivities of the binarization to the chosen
parameters. 2.1 Primary human hepatocytes 2.2 HepG2 cells
(the horizontal line indicates the first (top) and the second (bottom)
measurement set for HepG2 cells); Parameter p1 (2.1a, 2.2a): the
ratio between the value at time 1 and the value at time 0 lies
beneath (red) or above (green) the fixed threshold p1 = 1.5;
Parameter p2 (2.1b, 2.2b): the ratio between the signal and the
maximum value for this signal from all measurements lies beneath
(red) or above (green) the fixed threshold p2 = 0.15; Parameter p3
(2.1c, 2.2c): the signal lies beneath (red) or above (green) the fixed
threshold for experimental noise (p3 = 100). For all parameters:
The darker a field is colored, the larger is the distance to the
chosen threshold, i.e. the binarization is less sensitive on the
parameter.
Found at: doi:10.1371/journal.pcbi.1000438.s002 (0.61 MB PDF)
Figure S3.
Comparison of the discretized data with predictions
from model M2. A Primary human hepatocytes (data from
Alexopoulos et al, in preparation). B HepG2 cells (the horizontal
line separates the the first (top) from the second (bottom) dataset
for HepG2 cells; see also text). Each row represents one treatment
and the readouts are shown in the columns. Light green: predicted
correctly, ‘‘on’’; dark green: predicted correctly, ‘‘off’’; light red:
predicted ‘‘on’’, measured ‘‘off’’; dark red: predicted ‘‘off’’,
measured ‘‘on’’; yellow: state cannot be determined in logical
steady state analysis; black: data points where the measured species
is inhibited are not considered.
Found at: doi:10.1371/journal.pcbi.1000438.s003 (0.21 MB PDF)
Figure S4.
Data plots generated with DataRail. Shown are the
phosphorylation states of the proteins after 0, 30 and 180 minutes.
Green: significant activation after 30 minutes (according to the
chosen parameters); gray: no significant activation (cf. also Saez-
Rodriguez et al, 2008). A Primary human hepatocytes (data
obtained from Alexopoulos et al (in preparation)) B HepG2 cells,
first set of experiments (data obtained from Alexopoulos et al (in
preparation)) C HepG2 cells, second set of experiments.
Found at: doi:10.1371/journal.pcbi.1000438.s004 (0.28 MB PDF)
Table S1. Logical EGFR/ErbB model: list of species and interactions.
Found at: doi:10.1371/journal.pcbi.1000438.s005 (0.16 MB PDF)
Table S2. Incomplete truth tables (ITTs) in the model variant M2.
Found at: doi:10.1371/journal.pcbi.1000438.s006 (0.01 MB PDF)
Table S3. Proposed model changes to improve the fit of the model to
the data.
Found at: doi:10.1371/journal.pcbi.1000438.s007 (0.01 MB PDF)
Acknowledgments
We thank Sebastian Mirschel for his support in building and visualizing the
network with ProMoT.
Author Contributions
Conceived and designed the experiments: JS-R LGA PKS. Performed the
experiments: LGA. Analyzed the data: RS JS-R LGA SK. Contributed
reagents/materials/analysis tools: RS JS-R LGA PKS SK. Wrote the
paper: RS JS-R LGA PKS SK. Software and algorithm development: RS.
Software and algorithm development: SK.
References
1. Olayioye MA, Neve RM, Lane HA, Hynes NE (2000) The ErbB signaling
network: receptor heterodimerization in development and cancer. EMBO J 19:
3159–3167.
2. Yarden Y, Sliwkowski MX (2001) Untangling the ErbB signalling network. Nat
Rev Mol Cell Biol 2: 127–137.
3. Wiley HS, Cunningham DD (1981) A steady state model for analyzing the
cellular binding, internalization and degradation of polypeptide ligands. Cell 25:
433–440.
4. Citri A, Yarden Y (2006) EGF-ERBB signalling: towards the systems level. Nat
Rev Mol Cell Biol 7: 505–516.
5. Wiley HS, Shvartsman SY, Lauffenburger DA (2003) Computational modeling
of the EGF-receptor system: a paradigm for systems biology. Trends Cell Biol
13: 43–50.
6. Wofsy C, Goldstein B, Lund K, Wiley HS (1992) Implications of epidermal
growth factor (EGF) induced egf receptor aggregation. Biophys J 63: 98–110.
7. Kholodenko BN, Demin OV, Moehren G, Hoek JB (1999) Quantification of
short term signaling by the epidermal growth factor receptor. J Biol Chem 274:
30169–30181.
8. Resat H, Ewald JA, Dixon DA, Wiley HS (2003) An integrated model of
epidermal growth factor receptor trafficking and signal transduction. Biophys J
85: 730–743.
9. Schoeberl B, Eichler-Jonsson C, Gilles ED, Muller G (2002) Computational
modeling of the dynamics of the MAP kinase cascade activated by surface and
internalized EGF receptors. Nat Biotechnol 20: 370–375.
10. Birtwistle MR, Hatakeyama M, Yumoto N, Ogunnaike BA, Hoek JB, et al.
(2007) Ligand-dependent responses of the ErbB signaling network: experimental
and modeling analyses. Mol Syst Biol 3: 144.
11. Hatakeyama M, Kimura S, Naka T, Kawasaki T, Yumoto N, et al. (2003) A
computational model on the modulation of mitogen-activated protein kinase
(MAPK) and Akt pathways in heregulin-induced ErbB signalling. Biochem J
373: 451–463.
12. Hendriks BS, Cook J, Burke JM, Beusmans JM, Lauffenburger DA, et al. (2006)
Computational modelling of ErbB family phosphorylation dynamics in response
to transforming growth factor alpha and heregulin indicates spatial compart-
mentation of phosphatase activity. Syst Biol (Stevenage) 153: 22–33.
13. Chen WW, Schoeberl B, Jasper PJ, Niepel M, Nielsen UB, et al. (2009) Input-
output behavior of ErbB signaling pathways as revealed by a mass action model
trained against dynamic data. Mol Syst Biol 5: 239.
14. Oda K, Matsuoka Y, Funahashi A, Kitano H (2005) A comprehensive pathway
map of epidermal growth factor receptor signaling. Mol Syst Biol 1: 2005 0010.
15. Binder B, Ebenhoh O, Hashimoto K, Heinrich R (2006) Expansion of signal
transduction networks. Syst Biol (Stevenage) 153: 364–368.
16. Ruths DA, Nakhleh L, Iyengar MS, Reddy SA, Ram PT (2006) Hypothesis
generation in signaling networks. J Comput Biol 13: 1546–1557.
17. Price ND, Reed JL, Palsson BO (2004) Genome-scale models of microbial cells:
evaluating the consequences of constraints. Nat Rev Microbiol 2: 886–897.
18. Jeong H, Mason SP, Barabasi AL, Oltvai ZN (2001) Lethality and centrality in
protein networks. Nature 411: 41–42.
19. Wagner A, Wright J (2004) Compactness and cycles in signal transduction and
transcriptional regulation networks: A signature of natural selection? Advances
in Complex Systems 7: 419–432.
20. Sackmann A, Heiner M, Koch I (2006) Application of Petri net based analysis
techniques to signal transduction pathways. BMC Bioinformatics 7: 482.
21. Zevedei-Oancea I, Schuster S (2005) A theoretical framework for detecting
signal transfer routes in signalling networks. Computers & Chemical Engineering
29: 597–617.
22. Papin JA, Palsson BO (2004) Topological analysis of mass-balanced signaling
networks: a framework to obtain network properties including crosstalk. Journal
of Theoretical Biology 227: 283–297.
23. Kauffman SA (1969) Metabolic stability and epigenesis in randomly constructed
genetic nets. J Theor Biol 22: 437–467.
24. Albert R, Othmer HG (2003) The topology of the regulatory interactions
predicts the expression pattern of the segment polarity genes in Drosophila
melanogaster. Journal of Theoretical Biology 223: 1–18.
25. Chaves M, Albert R, Sontag ED (2005) Robustness and fragility of Boolean
models for genetic regulatory networks. J Theor Biol 235: 431–449.
26. Mendoza L, Thieffry D, Alvarez-Buylla ER (1999) Genetic control of flower
morphogenesis in Arabidopsis thaliana: a logical analysis. Bioinformatics 15:
593–606.
27. Thomas R, D’Ari R (1990) Biological feedback. Boca RatonFlorida: CRC Press.
28. Saez-Rodriguez J, Simeoni L, Lindquist JA, Hemenway R, Bommhardt U, et al.
(2007) A logical model provides insights into T cell receptor signaling. PLoS
Comput Biol 3: e163.
29. Helikar T, Konvalina J, Heidel J, Rogers JA (2008) Emergent decision-making
in biological signal transduction networks. Proc Natl Acad Sci U S A 105:
1913–1918.
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
18
August 2009 | Volume 5 | Issue 8 | e1000438
30. Klamt S, Saez-Rodriguez J, Lindquist JA, Simeoni L, Gilles ED (2006) A
methodology for the structural and functional analysis of signaling and
regulatory networks. BMC Bioinformatics 7: 56.
31. Saez-Rodriguez J, Mirschel S, Hemenway R, Klamt S, Gilles ED, et al. (2006)
Visual setup of logical models of signaling and regulatory networks with
ProMoT. BMC Bioinformatics 7: 506.
32. Klamt S, Saez-Rodriguez J, Gilles ED (2007) Structural and functional analysis
of cellular networks with CellNetAnalyzer. BMC Syst Biol 1: 2.
33. Thieffry D (2007) Dynamical roles of biological regulatory circuits. Brief
Bioinform 8: 220–225.
34. Thomas R, Kaufman M (2001) Multistationarity, the basis of cell differentiation
and memory. I. Structural conditions of multistationarity and other nontrivial
behavior. Chaos 11: 170–179.
35. Rodrigues GA, Falasca M, Zhang Z, Ong SH, Schlessinger J (2000) A novel
positive feedback loop mediated by the docking protein Gab1 and phosphati-
dylinositol 3-kinase in epidermal growth factor receptor signaling. Mol Cell Biol
20: 1448–1459.
36. Downward J (1998) Ras signalling and apoptosis. Curr Opin Genet Dev 8: 49–54.
37. Bos JL (1989) ras oncogenes in human cancer: a review. Cancer Res 49: 4682–4689.
38. Burgard AP, Nikolaev EV, Schilling CH, Maranas CD (2004) Flux coupling
analysis of genome-scale metabolic network reconstructions. Genome Res 14:
301–312.
39. Pfeiffer T, Sanchez-Valdenebro I, Nuno JC, Montero F, Schuster S (1999)
METATOOL: for studying metabolic networks. Bioinformatics 15: 251–257.
40. Gagneur J, Klamt S (2004) Computation of elementary modes: a unifying
framework and the new binary approach. BMC Bioinformatics 5: 175.
41. Carver RS, Stevenson MC, Scheving LA, Russell WE (2002) Diverse expression
of ErbB receptor proteins during rat liver development and regeneration.
Gastroenterology 123: 2017–2027.
42. Saez-Rodriguez J, Goldsipe A, Muhlich J, Alexopoulos LG, Millard B, et al.
(2008) Flexible informatics for linking experimental data to mathematical models
via DataRail. Bioinformatics 24: 840–847.
43. Olayioye MA, Beuvink I, Horsch K, Daly JM, Hynes NE (1999) ErbB receptor-
induced activation of stat transcription factors is mediated by Src tyrosine
kinases. J Biol Chem 274: 17209–17218.
44. Shoyab M, Plowman GD, McDonald VL, Bradley JG, Todaro GJ (1989)
Structure and function of human amphiregulin: a member of the epidermal
growth factor family. Science 243: 1074–1076.
45. Hou Z, He L, Qi RZ (2007) Regulation of s6 kinase 1 activation by
phosphorylation at ser-411. J Biol Chem 282: 6922–6928.
46. Weng QP, Kozlowski M, Belham C, Zhang A, Comb MJ, et al. (1998)
Regulation of the p70 S6 kinase by phosphorylation in vivo. Analysis using site-
specific anti-phosphopeptide antibodies. J Biol Chem 273: 16621–16629.
47. Grimes CA, Jope RS (2001) The multifaceted roles of glycogen synthase kinase 3
beta in cellular signaling. Progress in Neurobiology 65: 391–426.
48. Nelander S, Wang W, Nilsson B, She QB, Pratilas C, et al. (2008) Models from
experiments: combinatorial drug perturbations of cancer cells. Mol Syst Biol 4:
216.
49. Conradi C, Saez-Rodriguez J, Gilles ED, Raisch J (2005) Using chemical
reaction network theory to discard a kinetic mechanism hypothesis. Syst Biol
(Stevenage) 152: 243–248.
50. Craciun G, Tang Y, Feinberg M (2006) Understanding bistability in complex
enzyme-driven reaction networks. Proc Natl Acad Sci U S A 103: 8697–8702.
51. Saez-Rodriguez J, Hammerle-Fickinger A, Dalal O, Klamt S, Gilles ED, et al.
(2008) Multistability of signal transduction motifs. IET Syst Biol 2: 80–93.
The Logic of ErbB Signaling
PLoS Computational Biology | www.ploscompbiol.org
19
August 2009 | Volume 5 | Issue 8 | e1000438
|
19662154
|
mkk4 = ( mlk3 ) OR ( mekk1 ) OR ( mekk4 )
mkk7 = ( mekk1 )
erbb12 = ( ( btc AND ( ( ( erbb1 AND erbb2 ) ) ) ) AND NOT ( shp1d ) ) OR ( ( tgfa AND ( ( ( erbb1 AND erbb2 ) ) ) ) AND NOT ( shp1d ) ) OR ( ( bir AND ( ( ( erbb1 AND erbb2 ) ) ) ) AND NOT ( shp1d ) ) OR ( ( hbegf AND ( ( ( erbb1 AND erbb2 ) ) ) ) AND NOT ( shp1d ) ) OR ( ( egf AND ( ( ( erbb1 AND erbb2 ) ) ) ) AND NOT ( shp1d ) ) OR ( ( epr AND ( ( ( erbb1 AND erbb2 ) ) ) ) AND NOT ( shp1d ) )
raf1 = ( ( ras AND ( ( ( csrc ) ) ) ) AND NOT ( aktd ) ) OR ( ( pak1 AND ( ( ( ras ) ) ) ) AND NOT ( aktd ) )
raccdc42 = ( vav2 ) OR ( sos1esp8e3b1 )
creb = ( p90rsk ) OR ( mk2 )
stat1 = ( erbb11 AND ( ( ( csrc ) ) ) )
mkk3 = ( mlk3 )
rntre = ( esp8r AND ( ( ( erbb11 ) ) ) )
mek12 = ( mekk1 ) OR ( raf1 )
pi3k = ( erbb13 AND ( ( ( pi3kr ) ) ) ) OR ( erbb34 AND ( ( ( pi3kr ) ) ) ) OR ( erbb23 AND ( ( ( pi3kr ) ) ) ) OR ( pi3kr AND ( ( ( gab1 ) ) ) ) OR ( ras AND ( ( ( pi3kr ) ) ) )
tsc1_tsc2 = NOT ( ( akt ) )
ship2d = ( ship2 )
vav2 = ( erbb11 AND ( ( ( pip3 ) ) ) ) OR ( pi34p2 AND ( ( ( erbb11 ) ) ) )
gab1 = ( pip3 ) OR ( erbb11 ) OR ( grb2 )
mtor_rap = ( rheb AND ( ( ( mtorr ) ) ) )
gsk3 = NOT ( ( p90rsk AND ( ( ( akt ) ) ) ) )
sos1 = ( ( sos1r AND ( ( ( grb2 ) ) ) ) AND NOT ( p90rskerk12d ) )
rheb = NOT ( ( tsc1_tsc2 ) )
ccbl = ( erbb11 )
erbb23 = ( btc AND ( ( ( erbb3 AND erbb2 ) ) ) ) OR ( nrg2b AND ( ( ( erbb3 AND erbb2 ) ) ) ) OR ( nrg1a AND ( ( ( erbb3 AND erbb2 ) ) ) ) OR ( bir AND ( ( ( erbb3 AND erbb2 ) ) ) ) OR ( epr AND ( ( ( erbb3 AND erbb2 ) ) ) ) OR ( nrg1b AND ( ( ( erbb3 AND erbb2 ) ) ) )
mekk1 = ( raccdc42 )
mlk3 = ( raccdc42 )
p70s6_2 = ( pdk1 AND ( ( ( mtor_rap AND p70s6_1 ) ) ) )
nck = ( erbb44 ) OR ( erbb11 ) OR ( erbb14 )
mkk6 = ( mlk3 )
pak1 = ( nck AND ( ( ( raccdc42 ) ) ) ) OR ( grb2 AND ( ( ( raccdc42 ) ) ) )
pro_apoptotic = ( bad )
erbb34 = ( ( nrg1a AND ( ( ( erbb3 AND erbb4 ) ) ) ) AND NOT ( erbb2 ) ) OR ( ( nrg2b AND ( ( ( erbb3 AND erbb4 ) ) ) ) AND NOT ( erbb2 ) ) OR ( ( nrg2a AND ( ( ( erbb3 AND erbb4 ) ) ) ) AND NOT ( erbb2 ) ) OR ( ( nrg1b AND ( ( ( erbb3 AND erbb4 ) ) ) ) AND NOT ( erbb2 ) )
mekk4 = ( raccdc42 )
shp1d = ( shp1 )
erk12 = ( mek12 )
limk1 = ( pak1 )
rab5a = ( ( rin1 ) AND NOT ( rntre ) )
elk1 = ( ( nucerk12 ) AND NOT ( pp2b ) )
pip3 = ( ( ( pi3k ) AND NOT ( ship2d ) ) AND NOT ( ptend ) )
erbb11 = ( ( ( btc AND ( ( ( NOT endocyt_degrad ) ) OR ( ( erbb1 ) ) ) ) AND NOT ( shp1d ) ) OR ( erbb1 AND ( ( ( NOT endocyt_degrad ) ) ) ) OR ( ( ar AND ( ( ( erbb1 ) ) OR ( ( NOT endocyt_degrad ) ) ) ) AND NOT ( shp1d ) ) OR ( ( egf AND ( ( ( erbb1 ) ) OR ( ( NOT endocyt_degrad ) ) ) ) AND NOT ( shp1d ) ) OR ( ( epr AND ( ( ( erbb1 ) ) OR ( ( NOT endocyt_degrad ) ) ) ) AND NOT ( shp1d ) ) OR ( ( tgfa AND ( ( ( NOT endocyt_degrad ) ) OR ( ( erbb1 ) ) ) ) AND NOT ( shp1d ) ) OR ( ( bir AND ( ( ( erbb1 ) ) OR ( ( NOT endocyt_degrad ) ) ) ) AND NOT ( shp1d ) ) OR ( shp1d AND ( ( ( NOT endocyt_degrad ) ) ) ) OR ( ( hbegf AND ( ( ( erbb1 ) ) OR ( ( NOT endocyt_degrad ) ) ) ) AND NOT ( shp1d ) ) ) OR NOT ( epr OR hbegf OR endocyt_degrad OR ar OR erbb1 OR shp1d OR tgfa OR egf OR btc OR bir )
erbb44 = ( btc AND ( ( ( erbb4 ) ) ) ) OR ( nrg2b AND ( ( ( erbb4 ) ) ) ) OR ( nrg1a AND ( ( ( erbb4 ) ) ) ) OR ( nrg4 AND ( ( ( erbb4 ) ) ) ) OR ( bir AND ( ( ( erbb4 ) ) ) ) OR ( nrg3 AND ( ( ( erbb4 ) ) ) ) OR ( nrg1b AND ( ( ( erbb4 ) ) ) )
pkc = ( pdk1 AND ( ( ( dag AND ca ) ) ) )
shc = ( erbb44 ) OR ( erbb11 ) OR ( erbb24 ) OR ( erbb13 ) OR ( erbb12 ) OR ( erbb23 ) OR ( erbb34 ) OR ( erbb14 )
ap1 = ( cfos AND ( ( ( cjun ) ) ) )
plcg = ( erbb11 )
erbb13 = ( ( ( nrg1a AND ( ( ( erbb3 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( tgfa AND ( ( ( erbb3 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( btc AND ( ( ( erbb3 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( nrg2a AND ( ( ( erbb3 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ar AND ( ( ( erbb3 AND erbb1 ) ) ) ) AND NOT ( shp1d ) ) OR ( ( ( egf AND ( ( ( erbb3 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( epr AND ( ( ( erbb3 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( nrg1b AND ( ( ( erbb3 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) )
stat5 = ( erbb24 AND ( ( ( csrc ) ) ) ) OR ( erbb11 AND ( ( ( csrc ) ) ) )
p90rskerk12d = ( p90rsk AND ( ( ( erk12 ) ) ) )
stat3 = ( erbb11 AND ( ( ( csrc ) ) ) )
aktd = ( akt )
p70s6_1 = ( jnk ) OR ( erk12 )
shp2 = ( gab1 )
cmyc = ( ( nucerk12 ) AND NOT ( gsk3 ) )
endocyt_degrad = ( ccbl AND ( ( ( rab5a ) ) ) )
grb2 = ( erbb11 ) OR ( erbb44 ) OR ( erbb13 ) OR ( erbb24 ) OR ( erbb23 ) OR ( erbb12 ) OR ( erbb34 ) OR ( erbb14 ) OR ( shc )
rin1 = ( ras )
cfos = ( ( p90rsk AND ( ( ( erk12 ) ) ) ) AND NOT ( pp2a ) ) OR ( ( jnk ) AND NOT ( pp2a ) )
akt = ( ( pdk1 AND ( ( ( pip3 AND mtor_ric ) ) ) ) AND NOT ( pp2a ) ) OR ( ( pi34p2 AND ( ( ( mtor_ric AND pdk1 ) ) ) ) AND NOT ( pp2a ) )
nucerk12 = ( ( erk12 ) AND NOT ( mkp ) )
mk2 = ( p38 )
ptend = ( pten )
rasgap = ( ( gab1 ) AND NOT ( shp2 ) )
actinreorg = ( limk1 )
p38 = ( mkk4 ) OR ( mkk3 ) OR ( mkk6 )
erbb24 = ( nrg1a AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( btc AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( nrg2b AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( nrg2a AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( nrg4 AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( nrg3 AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( egf AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( epr AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( nrg1b AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( tgfa AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( bir AND ( ( ( erbb4 AND erbb2 ) ) ) ) OR ( hbegf AND ( ( ( erbb4 AND erbb2 ) ) ) )
dag = ( plcg )
bad = NOT ( ( pak1 AND ( ( ( akt ) ) ) ) )
ip3 = ( plcg )
cjun = ( jnk )
p90rsk = ( erk12 AND ( ( ( pdk1 ) ) ) )
mtor_ric = ( mtorr )
jnk = ( mkk7 AND ( ( ( mkk4 ) ) ) )
sos1esp8e3b1 = ( sos1r AND ( ( ( pi3kr AND pip3 AND esp8r ) ) ) )
hsp27 = ( mk2 )
shp1 = ( erbb11 )
pi34p2 = ( ( ship2d AND ( ( ( pi3k ) ) ) ) AND NOT ( ptend ) )
ca = ( ip3 )
erbb14 = ( ( ( nrg2b AND ( ( ( erbb4 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( nrg1a AND ( ( ( erbb4 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( tgfa AND ( ( ( erbb4 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( nrg2a AND ( ( ( erbb4 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( nrg4 AND ( ( ( erbb4 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( egf AND ( ( ( erbb4 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( epr AND ( ( ( erbb4 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) ) OR ( ( ( nrg1b AND ( ( ( erbb4 AND erbb1 ) ) ) ) AND NOT ( erbb2 ) ) AND NOT ( shp1d ) )
ras = ( ( sos1 ) AND NOT ( rasgap ) )
|
Mathematical Modelling of Cell-Fate Decision in
Response to Death Receptor Engagement
Laurence Calzone1,2,3*, Laurent Tournier1,2,3, Simon Fourquet1,2,3, Denis Thieffry4,5, Boris Zhivotovsky6,
Emmanuel Barillot1,2,3", Andrei Zinovyev1,2,3"
1 Institut Curie, Paris, France, 2 Ecole des Mines ParisTech, Paris, France, 3 INSERM U900, Paris, France, 4 TAGC – INSERM U928 & Universite´ de la Me´diterrane´e, Marseille,
France, 5 CONTRAINTES Project, INRIA Paris-Rocquencourt, France, 6 Karolinska Institutet, Stockholm, Sweden
Abstract
Cytokines such as TNF and FASL can trigger death or survival depending on cell lines and cellular conditions. The
mechanistic details of how a cell chooses among these cell fates are still unclear. The understanding of these processes is
important since they are altered in many diseases, including cancer and AIDS. Using a discrete modelling formalism, we
present a mathematical model of cell fate decision recapitulating and integrating the most consistent facts extracted from
the literature. This model provides a generic high-level view of the interplays between NFkB pro-survival pathway, RIP1-
dependent necrosis, and the apoptosis pathway in response to death receptor-mediated signals. Wild type simulations
demonstrate robust segregation of cellular responses to receptor engagement. Model simulations recapitulate documented
phenotypes of protein knockdowns and enable the prediction of the effects of novel knockdowns. In silico experiments
simulate the outcomes following ligand removal at different stages, and suggest experimental approaches to further
validate and specialise the model for particular cell types. We also propose a reduced conceptual model implementing the
logic of the decision process. This analysis gives specific predictions regarding cross-talks between the three pathways, as
well as the transient role of RIP1 protein in necrosis, and confirms the phenotypes of novel perturbations. Our wild type and
mutant simulations provide novel insights to restore apoptosis in defective cells. The model analysis expands our
understanding of how cell fate decision is made. Moreover, our current model can be used to assess contradictory or
controversial data from the literature. Ultimately, it constitutes a valuable reasoning tool to delineate novel experiments.
Citation: Calzone L, Tournier L, Fourquet S, Thieffry D, Zhivotovsky B, et al. (2010) Mathematical Modelling of Cell-Fate Decision in Response to Death Receptor
Engagement. PLoS Comput Biol 6(3): e1000702. doi:10.1371/journal.pcbi.1000702
Editor: Rama Ranganathan, UT Southwestern Medical Center, United States of America
Received August 25, 2009; Accepted February 2, 2010; Published March 5, 2010
Copyright: 2010 Calzone et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work is supported by the APO-SYS EU FP7 project. LC, LT, SF, EB and AZ are members of the team ‘‘Systems Biology of Cancer,’’ Equipe labellise´e
par la Ligue Nationale Contre le Cancer. The study was also funded by the Projet Incitatif Collaboratif ‘‘Bioinformatics and Biostatistics of Cancer’’ at Institut Curie.
Work in BZ’s laboratory is supported by the Swedish and Stockholm Cancer Societies, the Swedish Childhood Cancer Foundation, the Swedish Research Council,
the EC-FP-6 (Oncodeath and Chemores) programs. DT acknowledges the support from the Belgian Federal Science Policy Office: IUAP P6/25 (BioMaGNet,
Bioinformatics and Modeling: from Genomes to Networks, 2007–2011). The funders had no role in study design, data collection and analysis, decision to publish,
or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: Laurence.Calzone@curie.fr
" These authors are joint senior authors on this work.
Introduction
Engagement of TNF or FAS receptors can trigger cell death by
apoptosis or necrosis, or yet lead to the activation of pro-survival
signalling pathway(s), such as NFkB. Apoptosis represents a tightly
controlled mechanism of cell death that is triggered by internal or
external death signals or stresses. This mechanism involves a
sequence of biochemical and morphological changes resulting in
the vacuolisation of cellular content, followed by its phagocyte-
mediated elimination. This physiological process regulates cell
homeostasis, development, and clearance of damaged, virus-
infected or cancer cells. In contrast, pathological necrosis results in
plasma membrane disruption and release of intracellular content
that can trigger inflammation in the neighbouring tissues. Long
seen as an accidental cell death, necrosis also appears regulated
and possibly involved in the clearance of virus-infected or cancer
cells that escaped apoptosis [1].
Dynamical modelling of the regulatory network controlling
apoptosis, non-apoptotic cell death and survival pathways could
help identify how and under which conditions the cell chooses
between different types of cellular deaths or survival. Moreover,
modelling could suggest ways to re-establish the apoptotic death
when it is altered, or yet to trigger necrosis in apoptosis-resistant
cells. The decision process involves several signalling pathways, as
well
as
multiple
positive
and
negative
regulatory
circuits.
Mathematical modelling provides a rigorous integrative approach
to understand and analyse the dynamical behaviours of such
complex systems.
Published models of cell death control usually focus on one
death pathway only, such as the apoptotic extrinsic or intrinsic
pathways [2,3,4]. A few studies integrate both pathways [5], some
show that the concentration of specific components contribute to
the decision between death and survival [6,7] while other studies
investigate the balance between proliferation, survival or apoptosis
in specific cell types along with the role of key components in these
pathways [8], but no mathematical models including necrosis are
available yet. Moreover, we still lack models properly demonstrat-
ing how cellular conditions determine the choice between necrosis,
PLoS Computational Biology | www.ploscompbiol.org
1
March 2010 | Volume 6 | Issue 3 | e1000702
apoptosis and survival, and how and to what extent conversions
are allowed between these fates.
Our study aims at identifying determinants of this cell fate
decision process. The three main phenotypes considered are
apoptosis, non-apoptotic cell death (which mainly covers necrosis) and
survival. Although the pathways leading to these three phenotypes
are highly intertwined, we first describe them separately hereafter,
concentrating on the players we chose to include in each pathway.
Summarised in Figure 1A, this description does not intend to be
exhaustive, but rather aims at covering the most established
processes participating in cell fate decision.
Caspase-dependent apoptotic cell death
Only the apoptotic caspase-dependent pathway downstream of
FAS and TNF receptors is considered here. Upon engagement by
their ligands and in the presence of FADD (FAS-Associated
protein with Death Domain), a specific Death Inducible Signalling
Complex (DISC-FAS or DISC-TNF in Figure 1) forms and
recruits pro-caspase-8. This leads to the cleavage and activation of
caspase-8 (CASP8). In the so-called type II cells, CASP8 triggers
the intrinsic or mitochondria-dependent apoptotic pathway, which
also responds to DNA damage directly through the p53-mediated
chain of events (not detailed here). CASP8 cleaves the BH3-only
protein BID (not explicitly included in the diagram), which can
then translocate to the mitochondria outer membrane. There,
BID competes with anti-apoptotic BH3 family members such as
BCL2 for interaction with the proteins BAX or BAK (BAX will
stand here for both BAX and BAK). Consequently, oligomerisa-
tion of BAX results in mitochondrial outer membrane permea-
bilisation (MOMP) and the release of pro-apoptotic factors. Once
released to the cytosol, cytochrome c (Cyt_c) interacts with
APAF1, recruiting pro-caspase-9. In presence of dATP, this
enables the assembly of the apoptosome complex (referred to as
‘Apoptosome’ in Figure 1A, lumping APAF1 and pro-caspase-9),
responsible for caspase-9 activation, followed by the proteolytic
activation of pro-caspase-3 (CASP3) [9]. By cleavage of specific
targets, the executioner caspases (CASP3 in the model) are
responsible for major biochemical and morphological changes
characteristic of apoptosis. SMAC/DIABLO (SMAC) is released
during MOMP to the cytosol, where it is able to inactivate the
caspase inhibitor XIAP [10]. CASP3 also participates in a positive
circuit by inducing the activation of CASP8 [11,12]. In type I cells,
CASP8 directly cleaves and activates executioner caspases such as
CASP3 (not described).
Non-apoptotic cell death (NonACD)
Here,
we
consider
mainly
a
mode
of
cell
death
with
morphological features of necrosis, which occurs when apoptosis
is impeded in cells treated with cytokines [13] or in some specific
cell lines such as L929 cells when exposed to TNF [14]. In primary
T cells, if caspases are inhibited, activation of TNFR or FAS
causes necrosis via a pathway that requires the protein RIP1 and
its kinase activity (RIP1K) [13]. This RIP1-dependent cytokine-
induced necrotic death defines necroptosis [15,16]. A genetic
screen recently identified other genes necessary for this type of cell
death [17]. However, a precise description of this pathway is still
lacking. Reactive oxygen species (ROS) were proposed to be
involved downstream of RIP1 [18]. ROS are also thought to play
a key role in the control of mitochondria permeability transition
(MPT), since they are produced by damaged mitochondria and
can oxidize mitochondrial components, thus favouring MPT
[19,20,21]. Furthermore, the role of mitochondria in necrosis is
highlighted through the involvement of MPT, which causes a fatal
drop in ATP level and leads to necrotic death. Indeed, MPT
results from the inhibition of ATP/ADP exchange at the level of
mitochondrial membranes, or from the inhibition of oxidative
phosphorylation decreasing cellular ATP level and causing energy
failure [21,22]. Although there is evidence that necrosis is also
triggered by TNF- and FAS-independent pathways, these are not
yet considered in this study. These pathways include, for example,
calpain-mediated
cleavage
of
AIF
followed
by
its
nuclear
translocation [23,24], or PARP-1-mediated NAD+ depletion
[24,25].
Survival pathway
NFkB represents a family of transcription factors that play a
central role in inflammation, immune response to infections and
cancer development [26]. The ubiquitination of RIP1 at lysine 63
by cIAP leads to the activation of IKK and ultimately that of
NFkB [27]. In different cell types, especially in tumour cell lines,
activation of NFkB inhibits TNF-induced cell death [28]. This
effect
is
mediated
by
NFkB
target
genes:
cFLIP
inhibits
recruitment of CASP8 by FADD [29]; anti-apoptotic BCL2
family members inhibit MOMP and MPT [30,31,32]; XIAP acts
as a caspase inhibitor [33]; and ferritin heavy chain [34] or
mitochondrial SOD2 [35] decrease ROS levels (these mechanisms
are represented in Figure 1A by a direct inhibitory arrow from
NFkB to ROS). For the sake of simplicity, other NFkB target
genes that are known to inhibit TNF-induced apoptosis are
provisionally omitted in the model (e.g., A20; cf. [36,37]).
Our goal here is to provide a simplified but yet rigorous model
of the mechanisms underlying cell fate selection in response to the
engagement of FAS and TNF receptors. We have proceeded in
several steps. First, we have assembled a regulatory network
covering the main experimental data. Species and interactions
were selected on the basis of an extensive literature search and
integrated in the form of a diagram or ‘‘regulatory graph’’. This
diagram is then translated into a dynamical model. Our analysis
initially focuses on the determination of the asymptotic properties
of the system for different conditions, which correspond to the
possible phenotypes that the model can account for. Next, we
Author Summary
Activation of death receptors (TNFR and Fas) can trigger
either survival or cell death according to the cell type and
the cellular conditions. In other words, the same signal can
have antagonist responses. On one hand, the cell can
survive by activating the NFkB signalling pathway. On the
other hand, it can die by apoptosis or necrosis. Apoptosis
is a suicide mechanism, i.e., an orchestrated way to disrupt
cellular components and pack them into specialized
vesicles that can be easily removed from the environment,
whereas necrosis is a type of death that involves release of
intracellular
components
in
the
surrounding
tissues,
possibly causing inflammatory response and severe injury.
We, biologists and theoreticians, have recapitulated and
integrated known biological data from the literature into
an influence diagram describing the molecular events
leading to each possible outcome. The diagram has been
translated into a dynamical Boolean model. Simulations of
wild type, mutant cells and drug treatments qualitatively
match current data, and predict several novel mutant
phenotypes, along with general characteristics of the cell
fate decision mechanism: transient activation of some key
proteins in necrosis, mutual inhibitory cross-talks between
the three pathways. Our model can further be used to
assess contradictory data and address specific biological
questions through in silico experiments.
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
2
March 2010 | Volume 6 | Issue 3 | e1000702
TNF
FASL
TNFR
DISC-TNF
FADD
DISC-FAS
RIP1
CASP8
RIP1ub
RIP1K
cFLIP
IKK
BAX
cIAP
MOMP
ATP
CASP3
NFkB
ROS
BCL2
Cyt_c
Apoptosome
SMAC
XIAP
Survival
NonACD
Apoptosis
MPT
TNF
FASL
RIP1
CASP8
cIAP
MOMP
ATP
CASP3
NFkB
ROS
Survival
NonACD
Apoptosis
MPT
A
B
Figure 1. Regulatory networks of cell-fate decision. (A) Master model: the molecular interactions between the main components intervening
in the three pathways are described as an influence graph leading to the three cell fates: survival, non-apoptotic cell death and apoptosis. Dashed
lines denote the pathway borders. (B) The corresponding reduced model.
doi:10.1371/journal.pcbi.1000702.g001
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
3
March 2010 | Volume 6 | Issue 3 | e1000702
analyse the different trajectories leading to each phenotype in the
wild type and mutant situations. As quantitative data are still
largely lacking for this system, we use a qualitative logical
formalism and its implementation in the GINsim software [38].
As we shall see, proper model analysis can assess where and when
cell fate decisions are made, provide novel insight concerning the
general structure of the network, in particular concerning the
occurrence of cross-talks between pathways, and predict novel
mutant phenotypes and component activity patterns.
Results
The information gathered in the literature has been integrated
into a regulatory graph (Figure 1A). Our selection of molecular
players (nodes of the graph) is based on our current understanding
of the molecular mechanisms of cell fate decision. Documented
positive or negative effects among pairs of components are repre-
sented by signed arcs (Figure 1). Each node or arc is annotated
and associated with bibliographical references in the model file, as
well as in the accompanying documentation (cf. supplementary
Text S1).
Our current model encompasses three main pathways (Figure 1A):
the activation of a caspase-dependent apoptosis pathway, the RIP1-
kinase-dependent pathway leading to necrosis, and the activation of
the transcription factor NFkB with pro-survival effects. Other
pathways involved in cell death, such as growth factor receptors or
other RTK (receptor tyrosine kinase), TLR (Toll-like receptor), and
MAPK signalling pathways have been provisionally left out.
We defined specific ‘‘markers’’ or ‘‘read-outs’’ of the three cell
fates. When caspase-3 is activated, the cells are considered to be
apoptotic; when MPT occurs and the level of ATP drops
dramatically, the cells enter non-apoptotic cell death; finally,
when NFkB is activated, we consider that cells survive. Active
survival is thus monitored here by the activation of NFkB
pathway, in accordance with many studies, as opposed to passive
survival, which occurs when no death signals are engaged. In
reality, other pathways can interact downstream of NFkB
activation, which can reinforce or shut off survival. For now,
passive survival will be referred to as the ‘naı¨ve’ state, i.e. the stable
states with none of the three pathways activated.
Cross-talks between the three pathways
As mentioned before, the pathways are highly intertwined
(Figure 1A). For instance, the survival pathway interacts with the
apoptotic pathway at different points: cFLIP inhibits CASP8;
BCL2 blocks mitochondria pore opening through inhibition of
BAX (and BAK, implicitly represented in our model); and XIAP
blocks the activity of both CASP9 in the apoptosome and CASP3.
Conversely, the apoptotic pathway negatively regulates NFkB
activity through the CASP8-mediated cleavage and inactivation of
RIP1 upstream of NFkB. Because RIP1 operates upstream of the
necrotic pathway, this regulation also impacts necrosis. Moreover,
for the apoptosome to form, dATP (or/and ATP) is (/are) needed.
Consequently, in our model, when necrosis occurs, ATP pro-
duction drops, terminating apoptosis. Regarding the influence of
the survival pathway on the necrotic one, NFkB tentatively
stimulates the production of anti-oxidants that shuts off ROS level.
Both the necrotic and the apoptotic pathways are able to interact
with the survival pathway through the action of cIAP1/2, referred
to as cIAP in our model. More precisely, cIAP1 and 2 are E3-
ubiquitin ligases that target RIP1 for K63-linked polyubiquitina-
tion. They are essential intermediates in the activation of NFkB
downstream of TNF receptor [27]. Some synthetic molecules that
mimic the N-terminal of SMAC IAP-interacting motif have been
shown to induce cIAP1/2 auto-ubiquitination and subsequent
proteasomal degradation, thus blocking TNF-dependent NFkB
activation [39,40]. Tentatively, mitochondrial permeabilization in
the apoptosis or necrosis pathways could block TNF-induced NFkB
activation through the release of SMAC into the cytosol thereby
causing the inhibition of c-IAP1/2. Initially, cIAP was not included
in the model, which led to discrepancies between model simulations
and published data. Indeed, in FADD or CASP8 deletion mutants,
our preliminary model predicted only survival (not shown), whereas
both necrotic and survival phenotypes were observed in experi-
ments in the presence of TNF or FAS [41,42,43]. The consideration
of the path MOMP)SMAC =|cIAP)NFkB enabled us to
eliminate the discrepancies, both necrotic and survival phenotypes
were then obtained in the simulations, although it does not preclude
other mechanisms.
Dynamical logical model of cell fate decision
To transform the static map shown in Figure 1A into a
dynamical model accounting for the different scenarios or set of
events leading to one of the three phenotypes, we have to define
proper dynamical rules. Since there is little reliable quantitative
information on reaction kinetics and cellular conditions leading to
one or another phenotype, these rules must be sufficiently flexible
to cover all possible scenarios following death receptor activation.
The nodes encompassed in the map represent different things:
simple biochemical components (receptors, ligands, proteins or
metabolites): TNF, FASL, TNFR, FADD, FLIP, CASP8, RIP1,
IKK, NFkB, cIAP, BCL2, BAX, Cyt_c, SMAC, ROS, XIAP,
CASP3, ATP); specific modified forms of proteins: RIP1K (active
RIP1 kinase), RIP1ub (K63-ubiquitinated RIP1); complexes of
proteins: DISC-TNF (corresponding to TRADD, TRAF2, FADD,
proCASP8), DISC-FAS (corresponding to FAS, FADD, pro-
CASP8), apoptosome; cellular processes: MPT (Mitochondrial
Permeability
Transition)
and
MOMP
(Mitochondria
Outer
Membrane Permeabilisation).
A Boolean variable is associated with each of these nodes, which
can take only two logical values: ‘‘0’’ (false), denoting the absence
or inactivity of the corresponding component, and ‘‘1’’ (true),
denoting its active state.
Furthermore, a logical rule (or function) is assigned to each
node, defining how the different inputs (incoming arrows) combine
to control its level of activation. For example, CASP8 can be
activated (its value is set to ‘‘1’’) by DISC-TNF or DISC-FAS, but
only in the absence of cFLIP protein. This can be encoded into a
logical rule as follows: (DISC-TNF OR DISC-FAS) AND NOT
cFLIP. Several nodes correspond to simple inputs (TNF, FASL
and FADD). Their initial values are kept fixed during most
simulations.
On the basis of the regulatory graph and the associated logical
rules, we then proceeded with the exploration of the dynamical
properties of our model. We first focused on the identification of
all stable states and on their biological interpretation. Then, we
investigated the reachability of these stable states for different
initial conditions, for both wild type and mutant cases. Details on
the computational methods used are provided in the Methods
section and in the supplementary Text S1. The logical model has
been
filed
in
the
BioModels
database
with
the
reference
MODEL0912180000.
Identification of stable states
Analysis of the cell fate decision model (Figure 1A) led to the
identification of the 27 stable states showed in Figure 2. These
stable states are the sole attractors of the system under the
asynchronous assumption (see Methods). They thus represent all
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
4
March 2010 | Volume 6 | Issue 3 | e1000702
possible cellular asymptotical states. In other words, whatever the
initial conditions, a wild type cell will end up in one of these states
if we wait long enough. A closer look reveals that several stable
states correspond to each cellular fate, with few differing (minor)
component values.
This consideration led us to address the following questions: (i)
Does a cluster structure exist in the distribution of internal stable
states of the network? (ii) If so, in these clusters, could the
corresponding states be interpreted as slightly different realisations
of the same cellular phenotype? (iii) What would be the char-
acteristic signature of each cluster (conserved values of variables
inside each cluster)? (iv) What is the number of independent
variables defining the internal stable states of the network?
Standard statistical methods and clustering algorithms are
applied to group stable states. Figure 3 displays a projection of the
internal (without inputs and outputs) stable values into the 2D
space defined by the first two principal components of the
corresponding distribution. The first two principal components
explain 52% and 20% of the total variation, respectively (Table
S1). The first principal component can be associated with the
activity of NFkB pathway, while the second is determined mainly
by ATP and MPT status. These factors do appear to determine
the principal (independent) degrees of freedom for the internal
state of the network. A typical trajectory starting from any set of
initial conditions will thus quickly converge to the region under the
influence of these three components.
The 2-D graph (Figure 3) reveals a striking separation of the
stable states into 4 clusters: one cluster (blue circles) devoid of
significant activity, which we call the ‘naı¨ve’ state; one cluster
(green rhombs) corresponding to survival, with NFkB pathway
activated; and two clusters corresponding to the two different
modalities of cell death, apoptosis (orange squares) and necrosis
(purple triangles). K-means clustering using Euclidean and L1
distance perfectly reproduces these groupings, demonstrating that
the compact groups easily distinguishable on the PCA plot indeed
represent well-separated clusters in the original multidimensional
space.
Some interesting conclusions and predictions can be drawn just
by looking at the values of each component in each phenotypical
group. For instance, in the necrotic (purple) stable states, when
FADD is present (i.e. normal wild type conditions), RIP1 is always
OFF and CASP8 ON, even though RIP1 is required and CASP8
is dispensable for necrosis to occur. This observation suggests a
transient activation of RIP1 protein when switching on the
necrotic pathway in response to death receptors. However,
inactivation or cleavage of RIP1 is not per se a prerequisite for
necrosis, nor is CASP8 activation. Indeed, for the mutant models
in which CASP8 activation is impaired, such as CASP8 or FADD
deletion, there exist necrotic attractors with RIP1 = 1 (not shown).
Our model thus predicts that TNF-induced necrosis could occur
despite
CASP8-mediated
cleavage
of
RIP1.
An
attractive
experimental model in which such a transient activation of RIP1
could be tested is the mouse fibrosarcoma cell line L929. Upon
TNF exposure, these cells die by necrosis [44] and they have a
functional CASP8 [45], which is cleaved during TNF-induced cell
death [46].
Since RIP1 controls both the activation of NFkB and the level
of ROS, the same transient behaviour could be expected for the
survival phenotype. However, this is not observed with our model,
as RIP1 = 1 in all survival (green) stable states. This can be
explained by the regulatory circuit involving RIP1 and NFkB,
which is not functional in necrosis. Indeed, when NFkB is active, it
can mediate the synthesis of cFLIP, an inhibitor of CASP8, itself
an inhibitor of RIP1. Moreover, RIP1 is part of the positive circuit
that keeps NFkB ON. The model thus suggests that a sustained
RIP1 activity is needed for survival. How could this hypothesis be
experimentally assessed? If an experiment would reveal that RIP1
is only transiently activated upon death receptor activation, while
NFkB remains activated, the model would be contradicted. In that
case, one would need to look for other components capable to
maintain NFkB active.
Model reduction and dynamical analysis
The stable state analysis described above provides a first validation
of the master model presented in Figure 1A. On this basis, we
performed a more detailed analysis of the dynamics of the system.
We investigated which cell fates (stable states) can be reached from
specific initial conditions. Given a set of reachable stable states, can
we say something about their relative ‘‘attractivity’’?
To avoid the combinatorial explosion of the number of states to
consider, we have reduced the number of components while
preserving the relevant dynamical properties of the master model
(Figure 1B). Details of how this reduction is performed are
provided in the Methods section. The resulting network en-
compasses 11 components. The corresponding Boolean rules are
listed in Table 1. The size of the transition graph (211 = 2048) is
now amenable to a detailed dynamical analysis. First, the set of
attractors of this reduced model is identified: 13 attractors are
obtained, which are all stable states matching those found for the
master model when the input variable FADD = 1. Recall that, in
Figure 2. Stable states of the master model. values 0 and 1 are
represented by empty and full circles, respectively.To compare these
stable states to those of the simplified model, the values of FADD = 0
need to be deleted since FADD is not explicitly presented in the
reduced model. The first six rows (with NonACD = 0, Apoptosis = 0,
Survival = 0) correspond to the ‘‘naı¨ve’’ state. The following five rows
(with NonACD = 0, Apoptosis = 0, Survival = 1) correspond to ‘survival’.
The following eight rows (with NonACD = 0, Apoptosis = 1, Survival = 0)
correspond to ‘apoptosis’. The last eight rows (with NonACD = 1,
Apoptosis = 0, Survival = 0) correspond to ‘necrosis’.
doi:10.1371/journal.pcbi.1000702.g002
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
5
March 2010 | Volume 6 | Issue 3 | e1000702
the master model, both values of FADD were considered leading
to 13 stable states with FADD = 1 and 14 stable states with
FADD = 0. Using the theoretical results presented in [47] (mainly
Theorem 1), we can conclude that the 13 stable states of Figure 2
are the only attractors of the master model when FADD = 1.
Logical definition of ‘‘mutants’’
Based on the reduced model defined in Figure 1B and Table 1,
we derived 15 model variants representing biologically plausible
perturbations. We will abusively use the term ‘‘mutant’’ to refer to
these variants, even though they do not all technically correspond
to mutations. For instance, the ‘‘z-VAD mutant’’ simulates the
effect of caspase inhibitor z-VAD-fmk. Each mutant simulation
consists in a local alteration of our reduced model, which can be
qualitatively compared with results reported in the literature.
In the Boolean framework, such alterations amount to force the
level of certain variables to zero in the case of a gene deletion, or to
one in the case of a component over-expression. As we are using
the reduced version of the master model, some perturbed
components may be hidden by the reduction process. In such
cases, we change the logical rules of their (possibly indirect) targets
to take into account their effects. Table 2 lists the 15 variants of the
model considered, along with the modified logical rules, the
expected effects on the phenotypes according to the literature, and
short descriptions of simulation results.
Computation of reachable attractors
The
references
provided
in
Table
2
cover
experiments
performed on different cell types and with different experimental
conditions. In contrast, our cell fate model represents mechanisms
of cell fate decision in a generic cell, qualitatively recapitulating a
wide variety of cellular contexts. Given a cellular system, its
response to the activation of death receptors is determined by the
logical rules. However, the generic model presented here considers
equally all possible contexts and regulatory combinations. To
evaluate the relative likelihood of having a particular response in a
randomly chosen cellular system, we count the relative number of
possible trajectories from the stimulated ‘naı¨ve’ state to a given
phenotype. This analysis gives an idea on what is possible or
forbidden in a ‘generic’ cell.
Using dedicated methods and software [48], the set of reachable
stable states is calculated, starting from selected physiological
initial conditions, for the wild type and mutant models. The
physiological state is defined by fixing the variables ATP and cIAP
Figure 3. Projection of the internal stable state values onto the first two principal components. Four clusters are formed: the ‘naı¨ve’
cluster (round light blue circles) at the center; the survival cluster (green rhombs) characterised by high level of NFkB and related (see Table S1 for
details); the apoptosis cluster (orange squares) characterised by high levels of MOMP, Cyt_c, SMAC and ATP; and the non-apoptotic cell death - or
necrosis - cluster (purple triangles) with high levels of MPT, ROS, MOMP, Cyt_c and SMAC. In the latter three clusters, there are three subgroups of
stable states which correspond to the different inputs of the system: top stable states correspond to high TNF and FAS signals, middle stable states
have either one or the other signal while the lower ones have no inputs. As for the naive cluster, the two sets of stable states differ by their cIAP value.
Inputs and outputs are not included in the stable state binary vector.
doi:10.1371/journal.pcbi.1000702.g003
Table 1. Logical rules associated with the wild type reduced
model.
Node
Logical update rule
TNF
( INPUT NODE)
FAS
( INPUT NODE)
RIP19
NOT C8 AND (TNF OR FAS)
NFkB9
(cIAP AND RIP1) AND NOT C3
C89
(TNF OR FAS OR C3) AND NOT NFkB
cIAP9
(NFkB OR cIAP) AND NOT MOMP
ATP9
NOT MPT
C39
ATP AND MOMP AND NOT NFkB
ROS9
NOT NFkB AND (RIP1 OR MPT)
MOMP9
MPT OR (C8 AND NOT NFkB)
MPT9
ROS AND NOT NFkB
doi:10.1371/journal.pcbi.1000702.t001
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
6
March 2010 | Volume 6 | Issue 3 | e1000702
to ‘‘1’’ and all the other ones to ‘‘0’’. Different combinations of
TNF and FASL are considered. The probability to reach each
phenotype is computed as a fraction of the paths in the graph that
link physiological initial conditions to each cell fate (Figure 4 for
reduced model and Figure S4 for master model).
As expected, the absence of TNF and FASL can only lead to the
‘naı¨ve’ state (except of course when caspase-8 or NFkB are over-
expressed, for obvious reasons). This means that the inputs (TNF
and FASL) are needed for the system to effectively trigger the
decision process. This was expected since intracellular death
signals are not yet taken into account in the model. When TNF = 1
(Figure 4, right panel), for the wild type system, we observe that
three outcomes or phenotypes are reachable from the initial
condition, with different probabilities: ,10% for necrosis, ,30%
for active survival and ,60% for apoptosis. Although these
probabilities cannot be directly compared with experimental
results, they become useful when comparing different variants of
the model. For instance, an increase (or decrease) of a phenotype
probability between the wild type and a particular mutant can be
interpreted
as
a
gain
(or
a
loss)
of
effectiveness
of
the
corresponding pathway in that mutant. Such qualitative observa-
tions can then be confronted with published experimental results,
which are summarized in the last column of Table 2.
In most cases, activation of FASL and TNF lead to similar
effects (not shown), except in the case of the FADD deletion mutant
(Figure 5). As expected, this mutant cannot lead to cell death when
FASL is ON. In contrast, necrosis is still possible in the presence of
TNF. Interestingly, TNF-induced apoptosis is expected to be
blocked [49] whereas the qualitative analysis shows that apoptosis
is actually reachable in the model. Nevertheless, the probability of
this phenotype is very low (around 0.61%), which means that very
few trajectories may lead to apoptosis and it would thus be difficult
to obtain the corresponding cellular context.
Variation of the duration of receptor activation and its
effects
In the reachability analysis presented above, the value of TNF
and FASL are kept constant and therefore always ON (or always
OFF) along all trajectories. These qualitative simulations are useful
to characterize the asymptotic behaviour of the system when the
death receptor is engaged for a sufficiently long time. The
principle of ‘ligand removal’ experiments consists in characterizing
the decision process when it is subject to a temporary pulse of
TNF. Here, time is intrinsically discrete, meaning that the
duration of TNF pulse denoted td is represented by an integer
Table 2. Description of the different ‘‘mutant’’ versions of the reduced model.
Name
Modified rules
Expected phenotypes
Qualitative results
Anti-oxidant
ROS9=(RIP1 OR MPT)
Prediction.
Suppression of NFkB anti-oxidant effect leads to no change
in the decision process.
APAF1 deletion
C39=0
APAF12/2 mouse thymocytes are not impaired in
FAS-mediated apoptosis ([71]).
Apoptosis disappears and replaced by the naı¨ve state.
Necrosis and survival are close to the wild type case situations.
BAX deletion
MOMP9= MPT
BAX deletion blocks FAS or TNF+CHX - induced
apoptosis in some cell lines, such as HCT116 [72].
BAX deletion prevents apoptosis.
BCL2 over-
expression
MOMP9= MPT
MPT9=0
FAS induces the activation of NFkB pathway [29].
As expected, the survival and naive attractors are preserved
while both death pathways are inhibited.
CASP8 deletion
C89=0
Caspase-8 deficient MEFs [41] or Jurkat cells [42]
are resistant to FAS-mediated apoptotic cell death.
Apoptosis disappears. Compared to the wild type, a slight
increase of necrosis is observed, while survival becomes the
main cell fate.
constitutively
activated CASP8
C89=1
Prediction.
Over-expression of caspase-8 leads to a loss of NFkB activation.
cFLIP deletion
C89=TNF OR FAS OR C3
cFLIP2/2 MEFs are highly sensitive to FASL and
TNF [73].
The increase of apoptosis is effectively observed in the cFLIP
mutant; furthermore survival can no longer be sustained.
cIAP deletion
cIAP9= 0
NFkB activation in response to TNF is blocked [53].
NFkB activation is impaired, and only the apoptotic and
necrotic attractors can be reached.
FADD deletion
C89=C3 AND NOT NFkB
RIP19=NOT C8 AND TNF
FADD2/2 mouse thymocytes are resistant to FAS
mediated apoptosis [74]. FADD2/2 MEFs are resistant
to FASL and TNF [75]. In Jurkat cells treated with
TNF+CHX, apoptosis is turned into necrosis [43].
FASL signalling is blocked and the ‘naı¨ve’ attractor is the only
reachable one. In response to TNF, apoptosis disappears.
NFkB deletion
NFkB9=0
TNF induces both apoptosis and necrosis in
NF-kB p652/2 cells [76] or in IKKb2/2 fibroblasts [35].
This mutant shows a strong increase of necrosis (to be related
with concomitant apoptosis/necrosis).
constitutively
active NFkB
NFkB9=1
Prediction.
Both death pathways are shut down in this mutant.
RIP1 deletion
RIP19=0
RIPK12/2 MEFs are hypersensitivity to TNF, no
TNF-induced NFkB activation, [77].
Both survival and necrosis states become unreachable.
The effect of RIP1 silencing leads to a complete loss of the
decision process (apoptosis becoming the only outcome).
XIAP deletion
C39=ATP AND MOMP
No effect on TNF-induced toxicity in XIAP2/2 MEFs [78].
Behaviour similar to wild type.
z-VAD
C39=0
FAS induced apoptosis is blocked, though cells can
undergo death by necrosis [79]. FAS activates NFkB [80].
Induction of autophagic cell death observed [81].
The simulation of z-VAD mutant is similar to the silencing of
caspase-8 (which implies that caspase-8 shut down in the
model seems to have some priority over caspase-3 shutdown).
z-VAD+RIP1
deletion
C39=0
C89=0
RIP19=0
Upon TNF cells treated with z-VAD-fmk that are
RIP-deficient cannot activate the NFkB pathway
anymore and die by necrosis [13].
The conjugated effect of RIP1 deletion and caspase inhibition
impedes the system to trigger any of the three pathways
(the ‘naı¨ve’ state becomes the only possible outcome).
doi:10.1371/journal.pcbi.1000702.t002
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
7
March 2010 | Volume 6 | Issue 3 | e1000702
number. In order to simulate each experiment, N trajectories were
generated, starting from the ‘‘physiological’’ condition with
TNF = 1. At time td, the value of TNF is forced to zero. The
probabilities to reach the different phenotypes are then calculated
as explained in the Methods section. The average probabilities,
over the N computed trajectories, are represented in Figure 6, for
the wild type and the 15 mutants.
The purpose of this study is to investigate the dynamics of all the
mutants and how they reach the various possible phenotypes for
different lengths of TNF pulses. It provides a measurable way to
assess the appearance or disappearance of certain phenotypes
upon TNF induction. The curves of Figure 6 allow to link
explicitly the graphs of Figure 4 when TNF is ON (right panels)
and OFF (left panels) with the subjacent dynamics.
Let us compare the wild type case and the deletion of cFLIP as
an example of how to read these graphs. For early events, the two
cases behave similarly as expected (up to event 3). As TNF pulse is
prolonged, the apoptotic phenotype becomes more and more
pronounced and strongly favoured over the survival one in the
cFLIP mutant as opposed to the wild type conditions. This leads to
the complete disappearance of survival in the mutant. This
observation reinforces the role of cFLIP in the control of the
apoptotic pathway.
With the ‘ligand removal’ experiment, we can evaluate the
number of steps, in the reduced model, that are needed for the cell
to decide on its fate after TNF exposure. For almost all mutants
and wild type case, the choice is made around step 4. This means
that, after this point, even if TNF is removed, the cell has already
committed to a specific fate.
One surprise arises from the non-monotonic behaviour of
mutants for which apoptosis is suppressed (APAF1, BAX, caspase-
8 and FADD deletions and z-VAD-fmk treatment), tentatively
indicating a competition between components of the survival and
necrotic pathways. Indeed several inhibitory cross-talks could
explain this behaviour. These mutants also indicate the existence
of an optimal TNF induction for which the maximum rate of
necrosis is achieved (around step 2 in the corresponding mutants of
Figure 6).
A compact conceptual model
To complete our study of cell fate decision, we reasoned on the
simplest model of cell fate that can be deduced from the master
model described above.
The purpose here was to further simplify the network to obtain
a formal representation of the logical core of the network. We have
selected three components to represent the three cellular fates:
NFkB for survival, MPT for necrosis and CASP3 for apoptosis.
Based on reduction techniques and on the identification of all
possible directed paths between these three components [50], a
three-node diagram was deduced from the master model.
In this compact model, each original path (including regulatory
circuit) is represented by an arc whose sign denotes the influence of
the source node on its target. All original paths and the
corresponding arcs are recorded in Table 3. In some ambiguous
cases (e.g. influence of MPT on CASP3 or of NFkB on MPT), the
decision on the sign of the influence is based on the Boolean rules
and not on the paths only. Indeed, two negative and one positive
paths link NFkB to MPT. Therefore, the sign of the arc depends
not only on the states of BCL2 and of ROS, both feeding onto
MPT, but also on the rule controlling MPT value. Since the
Figure 4. Reachability of phenotypes starting from ‘‘physiological’’ initial conditions. The colours correspond to the phenotypes as
identified by the clustering algorithm (blue: ‘‘naı¨ve’’ survival state; green: survival through NFkB pathway; orange: apoptosis; purple: necrosis). Left
panel: TNF = FAS = 0, right panel: TNF = 1 and FAS = 0.
doi:10.1371/journal.pcbi.1000702.g004
NO INPUT
TNF=1, FAS=0
FAS=1, TNF=0
Necrosis
Survival
Naive
Naive
Apoptosis
Figure 5. FADD mutant breaks the symmetry of TNF and FAS-
induced pathways. Activation of death receptors in FADD deletion
mutant leads to different cell fates depending on the values of TNF and
FAS.
doi:10.1371/journal.pcbi.1000702.g005
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
8
March 2010 | Volume 6 | Issue 3 | e1000702
absence of BCL2 and the presence of ROS (Boolean ‘AND’ gate)
participate in the activation of MPT, if BCL2 is active, then MPT
is set to 0, even when ROS is ON. By extension, if NFkB is ON,
then MPT is 0, justifying the choice for a negative influence. In the
case of mutations eliminating all the negative influences, however,
a positive arrow must be considered.
The resulting molecular network is symmetrical: each node is
self-activating and is negatively regulated by the other nodes
(Figure 7, upper left panel). This is a conceptual picture re-
presenting the general architecture of the master model that can
help address specific questions. Even for this relatively simple
regulatory graph, there is a finite but quite high number of possible
logical rules. For now, we use a simple generic rule involving the
AND and NOT operators. For example, the logical rule for
CASP3 is: NOT MPT AND NOT NFkB AND CASP3. This
compact model has four stable states, each corresponding to one
Figure 6. Ligand removal experiments. The x-axis represents the (discrete) duration of the TNF pulse td (see text). At each discrete time point
along the x-axis, the TNF signal is turned off. The different curves represent the average probabilities to reach the different attractors after the pulse
(the number of trajectories N = 2000). Curves are coloured in blue for naı¨ve state, green for NFkB survival, orange for apoptosis, and purple for
necrosis.
doi:10.1371/journal.pcbi.1000702.g006
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
9
March 2010 | Volume 6 | Issue 3 | e1000702
cell fate, along with the ‘naı¨ve’ state (Figure 7, upper right panel).
This is coherent with what was observed from the analysis of the
complete model.
To validate our compact model, we verified that the simulations
of known mutations correspond to the published observations.
Here, when a hidden component is deleted, all the paths traversing
this component in the original graph are broken. If all the paths
corresponding to an arc of the compact model happen to be
broken, then it is removed. In the case of auto-regulation, not only
the link is broken but the node is also set to zero to avoid the node
to become active in the absence of death receptor activation.
Let us consider the CASP8 deletion mutant to illustrate this
approach (Figure 7, middle panels). For this mutant, several
arrows in the compact model have to be deleted. For example, the
arcs CASP3)CASP3 (paths 4+5 in Table 3) and CASP3xMPT
(path 17) clearly depend on the activation of CASP8. Note,
however, that CASP8 intervenes in other paths, which do not fully
rely on its sole activity. In the case of the arc NFkB)NFkB,
CASP8 depletion interrupts path 3, while path 2 can still enable
the NFkB auto-regulation. Consistent with the results from the
previous section, CASP8 depletion leads to the loss of the
apoptotic fate while the ‘naı¨ve’ stable state cannot be attained.
At this point, one could wonder how apoptosis could be re-
established in a CASP8 mutant. The analysis of the broken paths
suggests some experiments to bypass CASP8 and undergo CASP3
activation. On the basis of path 4, BAX, MOMP, SMAC and
XIAP are identified as potential targets, while path 5 points to
cytochrome c and apoptosome. One way to experimentally assess
this possibility would be to inject exogenous cytochrome c as it was
done in ‘wild type’ conditions [51], or yet provoke its release from
the mitochondria by forcing the opening of the pores. This is
possible only in the absence (or with low activity) of NFkB and in
the presence of ATP. Again, since no quantitative information can
be deduced from the path analysis proposed in this study, no
prediction can be made on the concentrations of proteins needed
to achieve a specific answer.
In a previous section, we postulated that an inhibition of the
survival pathway by the necrotic pathway is necessary to
reproduce some mutant phenotype. We suggested that cIAP could
play this role. Let us now test this hypothesis with our conceptual
model. We build the corresponding 3-node model without cIAP.
In the current version, cIAP plays two important roles, first as a
mandatory
intermediate
in
the
inhibitory
effect
of
MPT
(associated with necrosis) onto NFkB (survival) (path 13), next as
an obligate intermediate in the self-activation of NFkB (path 2).
The simulation (Figure 7, lower right panel) shows that in the
absence of cIAP, it is impossible to obtain the necrosis cell fate in
the CASP8 (and FADD) mutant(s), in agreement with our previous
conclusions and in support of our suggestion. A complete list of all
possible gene knockouts is provided in the Table S2.
This conceptual model analysis underlines the importance to
simplify in order to better understand the general structure of the
network and reason on it. Indeed, the simple 3-node network
enables us to grasp global functional aspects and propose specific
qualitative predictions.
Discussion
Mathematical models provide a way to test biological hypotheses
in silico. They recapitulate consistent heterogeneous published results
and assemble disseminated information into a coherent picture using
a coherent mathematical formalism (discrete, continuous, stochastic,
Table 3. List of paths corresponding to single arcs in the conceptual model of Figure 7.
Arc type
Arc
Paths on the regulatory graph
Sign of
regulation
Feedback circuits
MPT)MPT
1)
MPT)ROS)MPT
(+)
NFkB)NFkB
2)
NFkB)cIAP)RIP1ub)IKK)NFkB
(+)
3)
NFkB)cFLIPxCASP8xRIP1)RIP1ub)IKK)NFkB
(+)
CASP3)CASP3
4)
CASP3)CASP8)BAX)MOMP)SMACxXIAPxCASP3
(+)
5)
CASP3)CASP8)BAX)MOMP)Cyt_c)apoptosome)CASP3
(+)
Other regulatory paths
CASP3xNFkB
6)
CASP3)CASP8xRIP1)RIP1ub)IKK)NFkB
(2)
7)
CASP3)CASP8)BAX)MOMP)SMACxcIAP)RIP1ub)IKK)NFkB
(2)
8)
CASP3xNFkB
(2)
NFkBxCASP3
9)
NFkB)cFLIPxCASP8)BAX)MOMP)Cyt_c)apoptosome)CASP3
(2)
10)
NFkB)XIAPxCASP3
(2)
11)
NFkB)XIAPxApoptosome)CASP3
(2)
12)
NFkB)BCL2xBAX)MOMP)Cyt_c)apoptosome)CASP3
(2)
MPTxNFkB
13)
MPT)MOMP)SMACxcIAP)RIP1ub)IKK)NFkB
(2)
NFkBxMPT
14)
NFkBxROS)MPT
(2)
15)
NFkB)BCL2xMPT
(2)
16)
NFkB)cFLIPxCASP8xRIP1)RIP1K)ROS)MPT
(+)
CASP3xMPT
17)
CASP3)CASP8xRIP1)RIP1K)ROS)MPT
(2)
MPTxCASP3
18)
MPT)MOMP)Cyt_c)apoptosome)CASP3
(+)
19)
MPT)MOMP)SMACxXIAPxCASP3
(+)
20)
MPT)MOMP)SMACxXIAPxapoptosome)CASP3
(+)
21)
MPTxATP)apoptosome)CASP3
(2)
doi:10.1371/journal.pcbi.1000702.t003
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
10
March 2010 | Volume 6 | Issue 3 | e1000702
hybrid, etc.), depending on the questions and the available data.
Then, modelling consists of constantly challenging the obtained
model with available published data or experimental results (mutants
or drug treatments). After several refinement rounds, a model
becomes particularly useful when it can provide counter-intuitive
insights or suggest novel promising experiments.
Here, we have conceived a mathematical model of cell fate
decision, based on a logical formalisation of well-characterised
molecular interactions. Former mathematical models only consid-
ered two cellular fates, apoptosis and cell survival. In contrast, we
include a non-apoptotic modality of cell death, mainly necrosis,
involving RIP1, ROS and mitochondria functions.
Both the master and the reduced models were constructed on
the basis of an extensive analysis of the literature. The master
model (Figure 1A) summarises our current understanding of the
mechanisms regulating cell fate decision and identifies the major
switches in this decision. However, some important interactions,
components (caspase-2, calpains, AIF, etc.) or pathways (JNK,
Akt, etc.) have not yet been considered. This model was built to
be as generic as possible. Most of the mutants considered were
analysed in Jurkat cells, T-cells, or L929 murine fibrosarcoma
cells, thus in very different cellular contexts (e.g. in response to
TNF, Jurkat cells are resistant to cell death, whereas L929 cell
lines undergo necrosis). We are trying to account for all those
phenotypes in a unique model. The next step will be to provide a
model variant for each cell type in order to better match cell-
specific behaviours.
The reduced models can be used to simulate observed
experiments and to reflect on the general mechanisms involved
in apoptosis, survival or necrosis. This led us to identify the
principal actors involved in the decision process. The presence of
RIP1 or FADD, for example, proved to be decisive in our
simulation. However, the role of cFLIP appears less obvious than
previously suggested [7].
We can easily perturb the structure of the system in silico and
assess the dynamical effects of such perturbations (e.g. novel
knockouts). Our model can also be used to decide between
antagonist results found in different publications. For instance, the
inhibitory role of cIAP1/2 on the apoptotic pathway was initially
attributed to a direct inhibition of caspases. However, detailed
biochemical studies challenged this view [52,53]. We have tested
this hypothesis by adding an inhibitory arc from cIAP onto
CASP8, but simulations do not support a functional inhibitory role
of cIAP1/2, since survival is favoured over apoptosis in many
CASP3
NFkB
MPT
CASP3
NFkB
MPT
CASP3
NFkB
MPT
111
011
101
110
010
100
001
000
111
001
101
110
100
010
000
011
111
001
101
110
010
011
000
100
Figure 7. Simplified view of the cell fate model structure. Left panels: compact regulatory graph deduced from the master model (top), along
with two variants (middle and low). Right panels: state transition graphs corresponding to each regulatory graph, using generic logical rules (cf. text).
Stable states are represented by ellipses (at the bottom of each state transition graph). Each stable state corresponds to one cell fate: 000 for the
‘naı¨ve’ state, 010 for survival, 001 for apoptosis, and 100 for necrosis. Top: wild type structure. Middle: CASP8 deletion mutant. Bottom: CASP8 deletion
mutant in the absence of cIAP.
doi:10.1371/journal.pcbi.1000702.g007
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
11
March 2010 | Volume 6 | Issue 3 | e1000702
mutants, thus making apoptosis a very improbable phenotype
(Figure S1). Similarly, we tested the role of the feedback circuit
involving CASP8 and CASP3. We found that the activation of
CASP8 by CASP3 is not functional when TNF and FASL are
constantly ON. However, when TNF or FAS signal is not
sustained, CASP3)CASP8 activation becomes necessary to
insure the persistence of the apoptotic phenotype. When TNF is
sustained, this feedback is no longer needed (see Figures S2 and S3
for details).
The in-depth analysis of model properties led us to propose
several predictions or novel insights. Some concern the structure of
the network, as several interactions appear to be necessary to
achieve specific phenotypes. For example, our simulations of
FADD and CASP8 deletion mutants underline the need for a
mechanism from the necrotic pathway that would inhibit the
survival one. Here, we consider a mechanism involving MPT,
SMAC and cIAP. Other simulations point to different roles of
proteins: RIP1 activity is transient in necrosis whereas it is
sustained in survival. Similarly, our model analysis shows the role
played by the duration of the TNF pulses in the cell fate decision
and enlights when this decision is made. Finally, some hints about
possible scenarios for forcing or restoring a phenotype in mutants
are provided.
Deregulations of the signalling pathways studied here can lead
to drastic and serious consequences. Hanahan and Weinberg
proposed that escape of apoptosis, together with other alterations
of cellular physiology, represents a necessary event in cancer
promotion and progression [54]. As a result, somatic mutations
leading to impaired apoptosis are expected to be associated with
cancer. In the cell fate model presented here, most nodes can be
classified as pro-apoptotic or anti-apoptotic according to the
results of ‘‘mutant’’ model simulations, which are correlated with
experimental results found in the literature. Genes classified as
pro-apoptotic in our model include caspases-8 and -3, APAF1 as
part of the apoptosome complex, cytochrome c (Cyt_c), BAX, and
SMAC. Anti-apoptotic genes encompass BCL2, cIAP1/2, XIAP,
cFLIP, and different genes involved in the NFkB pathway,
including NFKB1, RELA, IKBKG and IKBKB (not explicit in the
model). Genetic alterations leading to loss of activity of pro-
apoptotic genes or to increased activity of anti-apoptotic genes
have been associated with various cancers. Thus, we can cross-list
the alterations of these genes deduced from the model with what is
reported in the literature and verify their role and implications in
cancer.
For
instance,
concerning
pro-apoptotic
genes,
frameshift
mutations in the ORF of the BAX gene are reported in .50%
of colorectal tumours of the micro-satellite mutator phenotype
[55]. Expression of CASP8 is reduced in ,24% of tumours from
patients with Ewing’s sarcoma [56]. Caspase-8 was suggested in
several studies to function as a tumour suppressor in neuroblas-
tomas [57] and in lung cancer [58].
On the other hand, constitutive activation of anti-apoptotic
genes is often observed in cancer cells. The most striking example
is the over-expression of the BCL2 oncogene in almost all follicular
lymphomas, which can result from a t(14;18) translocation that
positions BCL2 in close proximity to enhancer elements of the
immunoglobulin heavy-chain locus [59]. As for the survival
pathway, elevated NFkB activity, resulting from different genetic
alterations or expression of the v-rel viral NFkB isoform, is
detected in multiple cancers, including lymphomas and breast
cancers [60]. An amplification of the genomic region 11q22 that
spans over the cIAP1 and cIAP2 genes is associated with lung
cancers [61], cervical cancer resistance to radiotherapy [62], and
oesophageal squamous cell carcinomas [63].
A better understanding of the pro- or anti-apoptotic roles of
these genes involved in various cancers and their interactions with
other pathways would set a ground for re-establishing a lost death
phenotype and identifying druggable targets. The cell fate model
proposed here is a first step in this direction.
In the future, we will consider additional signalling cascades and
their cross-talks, following the path open by other groups [64]. In
parallel, we are contemplating the inclusion of other modalities of
cell death such as autophagy [65], which inhibits apoptosis
through BCL2 and is itself inhibited by apoptosis through Beclin1.
The functioning of the intrinsic apoptotic pathway and the
internal cellular mechanisms capable of triggering it could be
investigated in more details, taking advantage of recent molecular
analyses [66,67]. Finally, when systematic quantitative data
regarding the decision between multiple cell fates will become
available, our qualitative model could be used to design more
quantitative models adapted to specific cellular systems in order to
predict the probability for a given cell to enter into a particular cell
fate depending on stimuli.
Methods
Boolean formalism, synchronous vs. asynchronous
strategy
The computation of trajectories in the state space consists in the
calculation of sequences of states where each member of the
sequence is a logical successor of the previous one. As we choose to use
Boolean variables to encode the 25-dimensional master model, the
state space is the set S = {0,1}25. Although finite, the size of this set is
huge (more than 33 millions states). Furthermore, in the discrete
framework, the mathematical definition of the trajectories assumes
an updating rule for the variables. Two main strategies are usually
considered to analyse discrete models of biological networks. The
first one consists in updating all variables simultaneously, at each
time step. This synchronous strategy [68] has the advantage to
generate simple determinist dynamics, each state having one and
only one successor. Drawing a directed arrow from each of the 225
states to its successor, one constructs the synchronous transition
graph, comprising all synchronous trajectories of the system. The
determinism of the synchronous transition graph is a very strong
property that poorly portrays the complexity of the biochemical
processes that are modelled (some processes are likely to occur faster
than others). The second strategy, which is used in this paper,
consists in considering that only one component is updated at each
time, implying that a state may have several successors [69].
More precisely, to compute the set of asynchronous successors
of a state x = (x1,…,xn)M{0,1}n, one has to follow the three steps: (1)
compute the state F(x) = (f1(x),…, fn(x)), where fi is the Boolean rule
of the ith variable (F(x) is thus the synchronous successor of x); (2)
select the indices i such that xi?fi(x) (those are the indices of the
variables that are liable to change when the system is in state x);
and (3) for all such indices i, the state (x1,…,fi(x),…,xn) is an
asynchronous successor of x.
According to this definition, in the asynchronous approach, no a
priori hypothesis is made on the order of the events: all possible
orders are considered, which is much more satisfying from a
modelling point of view, as it is very difficult to know the relative
speeds of the different processes involved in the master model.
Note that the stable states of the model are independent on the
choice of the strategy (synchronous or asynchronous). Therefore,
the first analysis (based on the clustering of stable states) is valid
regardless the updating strategy.
Drawing an arrow from each state to its asynchronous
successors leads to the construction of an asynchronous transition
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
12
March 2010 | Volume 6 | Issue 3 | e1000702
graph, which comprises all possible asynchronous trajectories of
the system. To each arrow starting from the same state is
associated an equal probability (see [70] for details). This is a
strong assumption, which is the main reason why the exact values
of computed probabilities (of the different phenotypes) should not
be compared to experimental data in a quantitative manner.
Nevertheless, the same assumption has been made for all model
variants (mutants and drug treatments), thereby allowing compar-
ative studies. A systematic method to assess the impact of the
probability distribution is a key point towards a finer quantitative
analysis (work in progress).
As pointed earlier, the size of the transition graph is exponential
with respect to the number of variables, which constitutes a first
obstacle to the dynamical analysis. A second difficulty resides in
the fact that the asynchronous graph is not deterministic, as each
vertex may have more than one successor, which, given the size of
the graph, makes the application of classical graph algorithms
computationally heavy.
Model reduction
We have used a model reduction technique specifically adapted
to discrete systems, which mainly consists in iteratively ‘‘hiding’’
some variables, while keeping track of underlying regulatory
processes [47]. The main dynamical properties of the master
model, including stable states and other attractors are conserved in
the reduced model. Thanks to the computation of the reduced
asynchronous transition graph, relevant qualitative dynamical
properties of the model can be compared to experimental results
for wild type and in different mutant cases.
To reduce the number of species in the master model, each
logical rule is considered. For each removed component, the
information contained in its rule is included in the rules of its
targets such that no effective regulation is lost.
Many intermediate components could easily be replaced by a
proper rewriting of the logical rules associated with their target
nodes. For example, IKK has only one input (RIP1ub) and one
output (NFkB). Since its role in our model merely consists in
transmitting the signal from RIP1ub to NFkB, it can be easily
replaced by a straightforward change in the logical rule associated
with NFkB (implementing a direct activation from RIP1ub
instead of IKK). We also relied on the results of the clustering of
stable states and their associations with biologically plausible
phenotypes to select the key components to keep in the reduced
model: NFkB is the principal survival actor, while caspases-3 and
-8, together with the mitochondrial membrane permeability
variables (MOMP and MPT), determine apoptotic and non-
apoptotic cell deaths.
Let us consider the example of the removal of BAX and BCL2
(Figure 1 A and B). The regulators (or inputs) of these variables are
NFkB for BCL2 and CASP8 for BAX while their regulating
targets (or outputs) are MPT for BCL2 and MOMP for BAX.
BCL2 is directly activated by NFkB, and has two targets: MPT
and BAX. Therefore, BCL2 removal is performed by replacing
BCL2 by NFkB into the rules of the two targets, leading to the two
new
logical
rules:
MPT9 = ROS
AND
NOT
NFkB
and
BAX9 = C8 AND NOT NFkB. Applying the same process to
remove BAX, one obtains the following new rule for MOMP:
MOMP9 = MPT OR (C8 AND NOT NFkB).
The variables MOMP and MPT have now as inputs the
variables NFkB and CASP8. One can see that, in spite of the
disappearance of variables BAX and BCL2, their regulating roles
are still indirectly coded in the reduced system, ensuring that no
‘‘logical interaction’’ of the master model (i.e. activation or
inhibition) is actually lost during the reduction process. Table S3
lists the variables of the master model that are removed to obtain
the reduced model.
Some hypotheses were made when reducing the model. First,
FADD is considered to be constantly ON in wild type simulations.
Second, since the two complexes TNFR and DISC-TNF have
been removed together with the input FADD, the two deaths
ligands TNF and L have the exact same action in the reduced
model. Indeed, we consider that, in response to FAS death
receptor engagement as well as that of TNF; the activations of
both the survival and necrotic pathways RIP1-dependent. In this
case, one could then merge these variables and consider only one
input that could be called ‘‘external death receptor’’. However, we
choose to keep the two variables TNF and FASL, in the FADD
deletion mutant, the phenotype differs for TNF and FAS signal:
actually, only for that mutant is the symmetry of TNF and FAS
broken.
Supporting Information
Figure S1
Simulations of the reduced cell fate model incre-
mented by the interaction cIAPxCASP8 with TNF = 1, FasL = 0.
Found at: doi:10.1371/journal.pcbi.1000702.s001 (0.10 MB PDF)
Figure S2
Simulations of the model after deletion of the
interaction CASP3)CASP8 from the model with TNF = 1,
FasL = 0.
Found at: doi:10.1371/journal.pcbi.1000702.s002 (0.11 MB PDF)
Figure S3
Ligand removal simulation for the model with the
deletion of the interaction CASP3)CASP8.
Found at: doi:10.1371/journal.pcbi.1000702.s003 (0.08 MB PDF)
Figure S4
Comparative simulations between master model and
reduced model.
Found at: doi:10.1371/journal.pcbi.1000702.s004 (0.13 MB PDF)
Table S1
Contribution of the different variables into the first
two principal components. The colour marks relatively large
contributions, positive in red and negative in green.
Found at: doi:10.1371/journal.pcbi.1000702.s005 (0.01 MB PDF)
Table S2
List of mutants simulated with the conceptual 3-node
model.
Found at: doi:10.1371/journal.pcbi.1000702.s006 (0.03 MB PDF)
Table S3
List of variables hidden from the master model to
generate the reduced model.
Found at: doi:10.1371/journal.pcbi.1000702.s007 (0.07 MB PDF)
Text S1
Supplementary Text includes 1) GINsim Report of the
Annotated Model and 2) Supplementary references
Found at: doi:10.1371/journal.pcbi.1000702.s008 (0.16 MB PDF)
Acknowledgments
We thank Luca Grieco and Brigitte Kahn-Perle`s for critical reading of the
manuscript and Thomas Fink for discussions around the conceptual model.
We further thank Markus Rehm for fruitful discussions and advice during
the construction of the model.
Author Contributions
Conceived and designed the experiments: LC LT SF DT BZ EB AZ.
Performed the experiments: LC LT SF DT BZ EB AZ. Analyzed the data:
LC LT SF DT BZ EB AZ. Contributed reagents/materials/analysis tools:
LC LT SF DT BZ EB AZ. Wrote the paper: LC LT SF DT BZ EB AZ.
Created, validated and proposed predictions of the model: LC LT SF DT
BZ AZ. Coordinated the project: EB AZ. Edited and finalized the paper:
LC. Developed the methodology for cell fate probability prediction: LT.
Provided GINsim assistance: DT.
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
13
March 2010 | Volume 6 | Issue 3 | e1000702
References
1. Li M, Beg AA (2000) Induction of necrotic-like cell death by tumor necrosis
factor alpha and caspase inhibitors: novel mechanism for killing virus-infected
cells. J Virol 74: 7470–7477.
2. Rehm M, Huber HJ, Hellwig CT, Anguissola S, Dussmann H, et al. (2009)
Dynamics of outer mitochondrial membrane permeabilization during apoptosis.
Cell Death Differ 16: 613–623.
3. Eissing T, Conzelmann H, Gilles ED, Allgower F, Bullinger E, et al. (2004)
Bistability analyses of a caspase activation model for receptor-induced apoptosis.
J Biol Chem 279: 36892–36897.
4. Legewie S, Bluthgen N, Herzel H (2006) Mathematical modeling identifies
inhibitors of apoptosis as mediators of positive feedback and bistability. PLoS
Comput Biol 2: e120.
5. Fussenegger M, Bailey JE, Varner J (2000) A mathematical model of caspase
function in apoptosis. Nat Biotechnol 18: 768–774.
6. Bentele M, Lavrik I, Ulrich M, Stosser S, Heermann DW, et al. (2004)
Mathematical modeling reveals threshold mechanism in CD95-induced
apoptosis. J Cell Biol 166: 839–851.
7. Lavrik IN, Golks A, Riess D, Bentele M, Eils R, et al. (2007) Analysis of CD95
threshold signaling: triggering of CD95 (FAS/APO-1) at low concentrations
primarily results in survival signaling. J Biol Chem 282: 13664–13671.
8. Zhang R, Shah MV, Yang J, Nyland SB, Liu X, et al. (2008) Network model of
survival signaling in large granular lymphocyte leukemia. Proc Natl Acad
Sci U S A 105: 16308–16313.
9. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, et al. (1997)
Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex
initiates an apoptotic protease cascade. Cell 91: 479–489.
10. Du C, Fang M, Li Y, Li L, Wang X (2000) Smac, a mitochondrial protein that
promotes cytochrome c-dependent caspase activation by eliminating IAP
inhibition. Cell 102: 33–42.
11. Slee EA, Harte MT, Kluck RM, Wolf BB, Casiano CA, et al. (1999) Ordering
the cytochrome c-initiated caspase cascade: hierarchical activation of caspases-2,
-3, -6, -7, -8, and -10 in a caspase-9-dependent manner. J Cell Biol 144:
281–292.
12. Cowling V, Downward J (2002) Caspase-6 is the direct activator of caspase-8 in
the cytochrome c-induced apoptosis pathway: absolute requirement for removal
of caspase-6 prodomain. Cell Death Differ 9: 1046–1056.
13. Holler N, Zaru R, Micheau O, Thome M, Attinger A, et al. (2000) Fas triggers
an alternative, caspase-8-independent cell death pathway using the kinase RIP as
effector molecule. Nat Immunol 1: 489–495.
14. Fiers W, Beyaert R, Boone E, Cornelis S, Declercq W, et al. (1995) TNF-
induced intracellular signaling leading to gene induction or to cytotoxicity by
necrosis or by apoptosis. J Inflamm 47: 67–75.
15. Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, et al. (2009)
Classification of cell death: recommendations of the Nomenclature Committee
on Cell Death 2009. Cell Death Differ 16: 3–11.
16. Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, et al. (2005) Chemical inhibitor
of nonapoptotic cell death with therapeutic potential for ischemic brain injury.
Nat Chem Biol 1: 112–119.
17. Hitomi J, Christofferson DE, Ng A, Yao J, Degterev A, et al. (2008)
Identification of a molecular signaling network that regulates a cellular necrotic
cell death pathway. Cell 135: 1311–1323.
18. Morgan MJ, Kim YS, Liu ZG (2008) TNFalpha and reactive oxygen species in
necrotic cell death. Cell Res 18: 343–349.
19. Kowaltowski AJ, Castilho RF, Vercesi AE (2001) Mitochondrial permeability
transition and oxidative stress. FEBS Lett 495: 12–15.
20. Andreyev AY, Kushnareva YE, Starkov AA (2005) Mitochondrial metabolism of
reactive oxygen species. Biochemistry (Mosc) 70: 200–214.
21. Orrenius S, Gogvadze V, Zhivotovsky B (2007) Mitochondrial oxidative stress:
implications for cell death. Annu Rev Pharmacol Toxicol 47: 143–183.
22. Kroemer G, Galluzzi L, Brenner C (2007) Mitochondrial membrane
permeabilization in cell death. Physiol Rev 87: 99–163.
23. Jurewicz A, Matysiak M, Tybor K, Kilianek L, Raine CS, et al. (2005) Tumour
necrosis factor-induced death of adult human oligodendrocytes is mediated by
apoptosis inducing factor. Brain 128: 2675–2688.
24. Moubarak RS, Yuste VJ, Artus C, Bouharrour A, Greer PA, et al. (2007)
Sequential activation of poly(ADP-ribose) polymerase 1, calpains, and Bax is
essential in apoptosis-inducing factor-mediated programmed necrosis. Mol Cell
Biol 27: 4844–4862.
25. Xu Y, Huang S, Liu ZG, Han J (2006) Poly(ADP-ribose) polymerase-1 signaling
to mitochondria in necrotic cell death requires RIP1/TRAF2-mediated JNK1
activation. J Biol Chem 281: 8788–8795.
26. Karin M (2006) Nuclear factor-kappaB in cancer development and progression.
Nature 441: 431–436.
27. Varfolomeev E, Goncharov T, Fedorova AV, Dynek JN, Zobel K, et al. (2008)
c-IAP1 and c-IAP2 are critical mediators of tumor necrosis factor alpha
(TNFalpha)-induced NF-kappaB activation. J Biol Chem 283: 24295–24299.
28. Liu ZG, Hsu H, Goeddel DV, Karin M (1996) Dissection of TNF receptor 1
effector functions: JNK activation is not linked to apoptosis while NF-kappaB
activation prevents cell death. Cell 87: 565–576.
29. Kreuz S, Siegmund D, Scheurich P, Wajant H (2001) NF-kappaB inducers
upregulate cFLIP, a cycloheximide-sensitive inhibitor of death receptor
signaling. Mol Cell Biol 21: 3964–3973.
30. Wang CY, Guttridge DC, Mayo MW, Baldwin AS, Jr. (1999) NF-kappaB
induces expression of the Bcl-2 homologue A1/Bfl-1 to preferentially suppress
chemotherapy-induced apoptosis. Mol Cell Biol 19: 5923–5929.
31. Catz SD, Johnson JL (2001) Transcriptional regulation of bcl-2 by nuclear factor
kappa B and its significance in prostate cancer. Oncogene 20: 7342–7351.
32. Chipuk JE, Green DR (2008) How do BCL-2 proteins induce mitochondrial
outer membrane permeabilization? Trends Cell Biol 18: 157–164.
33. Turner DJ, Alaish SM, Zou T, Rao JN, Wang JY, et al. (2007) Bile salts induce
resistance to apoptosis through NF-kappaB-mediated XIAP expression. Ann
Surg 245: 415–425.
34. Pham CG, Bubici C, Zazzeroni F, Papa S, Jones J, et al. (2004) Ferritin heavy
chain upregulation by NF-kappaB inhibits TNFalpha-induced apoptosis by
suppressing reactive oxygen species. Cell 119: 529–542.
35. Kamata H, Honda S, Maeda S, Chang L, Hirata H, et al. (2005) Reactive
oxygen species promote TNFalpha-induced death and sustained JNK activation
by inhibiting MAP kinase phosphatases. Cell 120: 649–661.
36. Krikos A, Laherty CD, Dixit VM (1992) Transcriptional activation of the tumor
necrosis factor alpha-inducible zinc finger protein, A20, is mediated by kappa B
elements. J Biol Chem 267: 17971–17976.
37. Daniel S, Arvelo MB, Patel VI, Longo CR, Shrikhande G, et al. (2004) A20
protects endothelial cells from TNF-, Fas-, and NK-mediated cell death by
inhibiting caspase 8 activation. Blood 104: 2376–2384.
38. Naldi A, Berenguier D, Faure A, Lopez F, Thieffry D, et al. (2009) Logical
modelling of regulatory networks with GINsim 2.3. Biosystems 97: 134–139.
39. Vince JE, Wong WW, Khan N, Feltham R, Chau D, et al. (2007) IAP
antagonists target cIAP1 to induce TNFalpha-dependent apoptosis. Cell 131:
682–693.
40. Varfolomeev E, Blankenship JW, Wayson SM, Fedorova AV, Kayagaki N, et al.
(2007) IAP antagonists induce autoubiquitination of c-IAPs, NF-kappaB
activation, and TNFalpha-dependent apoptosis. Cell 131: 669–681.
41. Varfolomeev EE, Schuchmann M, Luria V, Chiannilkulchai N, Beckmann JS,
et al. (1998) Targeted disruption of the mouse Caspase 8 gene ablates cell death
induction by the TNF receptors, Fas/Apo1, and DR3 and is lethal prenatally.
Immunity 9: 267–276.
42. Kawahara A, Ohsawa Y, Matsumura H, Uchiyama Y, Nagata S (1998)
Caspase-independent cell killing by Fas-associated protein with death domain.
J Cell Biol 143: 1353–1360.
43. Harper N, Hughes M, MacFarlane M, Cohen GM (2003) Fas-associated death
domain protein and caspase-8 are not recruited to the tumor necrosis factor
receptor 1 signaling complex during tumor necrosis factor-induced apoptosis.
J Biol Chem 278: 25534–25541.
44. Krysko DV, Vanden Berghe T, D’Herde K, Vandenabeele P (2008) Apoptosis
and necrosis: detection, discrimination and phagocytosis. Methods 44: 205–221.
45. Yu L, Wan F, Dutta S, Welsh S, Liu Z, et al. (2006) Autophagic programmed
cell death by selective catalase degradation. Proc Natl Acad Sci U S A 103:
4952–4957.
46. Kouroku Y, Fujita E, Jimbo A, Mukasa T, Tsuru T, et al. (2000) Localization of
active form of caspase-8 in mouse L929 cells induced by TNF treatment and
polyglutamine aggregates. Biochem Biophys Res Commun 270: 972–977.
47. Naldi A, Remy E, Thieffry D, Chaouiya C (2009) A reduction method for logical
regulatory graphs preserving essential dynamical properties. Lect Notes Comput
Sci 5688: 266–280.
48. Tournier L, Chaves M (2009) Uncovering operational interactions in genetic
networks using asynchronous Boolean dynamics. J Theor Biol 260: 196–209.
49. Vanden Berghe T, van Loo G, Saelens X, Van Gurp M, Brouckaert G, et al.
(2004) Differential signaling to apoptotic and necrotic cell death by Fas-
associated death domain protein FADD. J Biol Chem 279: 7925–7933.
50. Zinovyev A, Viara E, Calzone L, Barillot E (2008) BiNoM: a Cytoscape plugin
for manipulating and analyzing biological networks. Bioinformatics 24: 876–877.
51. Zhivotovsky B, Orrenius S, Brustugun OT, Doskeland SO (1998) Injected
cytochrome c induces apoptosis. Nature 391: 449–450.
52. Eckelman BP, Salvesen GS, Scott FL (2006) Human inhibitor of apoptosis
proteins: why XIAP is the black sheep of the family. EMBO Rep 7: 988–994.
53. Varfolomeev E, Vucic D (2008) (Un)expected roles of c-IAPs in apoptotic and
NFkappaB signaling pathways. Cell Cycle 7: 1511–1521.
54. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100: 57–70.
55. Rampino N, Yamamoto H, Ionov Y, Li Y, Sawai H, et al. (1997) Somatic
frameshift mutations in the BAX gene in colon cancers of the microsatellite
mutator phenotype. Science 275: 967–969.
56. Lissat A, Vraetz T, Tsokos M, Klein R, Braun M, et al. (2007) Interferon-
gamma sensitizes resistant Ewing’s sarcoma cells to tumor necrosis factor
apoptosis-inducing ligand-induced apoptosis by up-regulation of caspase-8
without altering chemosensitivity. Am J Pathol 170: 1917–1930.
57. Teitz T, Lahti JM, Kidd VJ (2001) Aggressive childhood neuroblastomas do not
express caspase-8: an important component of programmed cell death. J Mol
Med 79: 428–436.
58. Shivapurkar N, Toyooka S, Eby MT, Huang CX, Sathyanarayana UG, et al.
(2002) Differential inactivation of caspase-8 in lung cancers. Cancer Biol Ther 1:
65–69.
59. Croce CM (2008) Oncogenes and cancer. N Engl J Med 358: 502–511.
60. Karin M, Cao Y, Greten FR, Li ZW (2002) NF-kappaB in cancer: from
innocent bystander to major culprit. Nat Rev Cancer 2: 301–310.
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
14
March 2010 | Volume 6 | Issue 3 | e1000702
61. Dai Z, Zhu WG, Morrison CD, Brena RM, Smiraglia DJ, et al. (2003) A
comprehensive search for DNA amplification in lung cancer identifies inhibitors
of apoptosis cIAP1 and cIAP2 as candidate oncogenes. Hum Mol Genet 12:
791–801.
62. Imoto I, Tsuda H, Hirasawa A, Miura M, Sakamoto M, et al. (2002) Expression
of cIAP1, a target for 11q22 amplification, correlates with resistance of cervical
cancers to radiotherapy. Cancer Res 62: 4860–4866.
63. Imoto I, Yang ZQ, Pimkhaokham A, Tsuda H, Shimada Y, et al. (2001)
Identification of cIAP1 as a candidate target gene within an amplicon at 11q22
in esophageal squamous cell carcinomas. Cancer Res 61: 6629–6634.
64. Aldridge BB, Saez-Rodriguez J, Muhlich JL, Sorger PK, Lauffenburger DA
(2009) Fuzzy logic analysis of kinase pathway crosstalk in TNF/EGF/insulin-
induced signaling. PLoS Comput Biol 5: e1000340.
65. Fimia GM, Piacentini M (2009) Toward the understanding of autophagy
regulation and its interplay with cell death pathways. Cell Death Differ 16:
933–934.
66. Krammer PH, Arnold R, Lavrik IN (2007) Life and death in peripheral T cells.
Nat Rev Immunol 7: 532–542.
67. Johnstone RW, Frew AJ, Smyth MJ (2008) The TRAIL apoptotic pathway in
cancer onset, progression and therapy. Nat Rev Cancer 8: 782–798.
68. Kauffman SA (1993) Origins of Order: Self- Organization and Selection in
Evolution Oxford University Press: N.Y.
69. Thomas R, Thieffry D, Kaufman M (1995) Dynamical behaviour of biological
regulatory networks–I. Biological role of feedback loops and practical use of the
concept of the loop-characteristic state. Bull Math Biol 57: 247–276.
70. Tournier L, Chaves M (2009) Uncovering operational interactions in genetic
networks using asynchronous Boolean dynamics. Journal of Theoretical Biology
260: 196–209.
71. Yoshida H, Kong YY, Yoshida R, Elia AJ, Hakem A, et al. (1998) Apaf1 is
required for mitochondrial pathways of apoptosis and brain development. Cell
94: 739–750.
72. LeBlanc H, Lawrence D, Varfolomeev E, Totpal K, Morlan J, et al. (2002)
Tumor-cell resistance to death receptor–induced apoptosis through mutational
inactivation of the proapoptotic Bcl-2 homolog Bax. Nat Med 8: 274–281.
73. Yeh WC, Itie A, Elia AJ, Ng M, Shu HB, et al. (2000) Requirement for Casper
(c-FLIP) in regulation of death receptor-induced apoptosis and embryonic
development. Immunity 12: 633–642.
74. Zhang J, Cado D, Chen A, Kabra NH, Winoto A (1998) Fas-mediated apoptosis
and activation-induced T-cell proliferation are defective in mice lacking FADD/
Mort1. Nature 392: 296–300.
75. Yeh WC, Pompa JL, McCurrach ME, Shu HB, Elia AJ, et al. (1998) FADD:
essential for embryo development and signaling from some, but not all, inducers
of apoptosis. Science 279: 1954–1958.
76. Sakon S, Xue X, Takekawa M, Sasazuki T, Okazaki T, et al. (2003) NF-kappaB
inhibits TNF-induced accumulation of ROS that mediate prolonged MAPK
activation and necrotic cell death. EMBO J 22: 3898–3909.
77. Kelliher MA, Grimm S, Ishida Y, Kuo F, Stanger BZ, et al. (1998) The death
domain kinase RIP mediates the TNF-induced NF-kappaB signal. Immunity 8:
297–303.
78. Harlin H, Reffey SB, Duckett CS, Lindsten T, Thompson CB (2001)
Characterization of XIAP-deficient mice. Mol Cell Biol 21: 3604–3608.
79. Kitanaka C, Kuchino Y (1999) Caspase-independent programmed cell death
with necrotic morphology. Cell Death Differ 6: 508–515.
80. Kreuz S, Siegmund D, Rumpf JJ, Samel D, Leverkus M, et al. (2004) NFkappaB
activation by Fas is mediated through FADD, caspase-8, and RIP and is
inhibited by FLIP. J Cell Biol 166: 369–380.
81. Vandenabeele P, Vanden Berghe T, Festjens N (2006) Caspase inhibitors
promote alternative cell death pathways. Sci STKE 2006: pe44.
Mathematical Model of Cell Fate Decision
PLoS Computational Biology | www.ploscompbiol.org
15
March 2010 | Volume 6 | Issue 3 | e1000702
|
20221256
|
RIP1 = ( ( DISC-FAS ) AND NOT ( CASP8 ) ) OR ( ( TNFR ) AND NOT ( CASP8 ) )
NonACD = ( NOT ( ( ATP ) ) ) OR NOT ( ATP )
XIAP = ( ( NFkB ) AND NOT ( SMAC ) )
NFkB = ( ( IKK ) AND NOT ( CASP3 ) )
RIP1ub = ( cIAP AND ( ( ( RIP1 ) ) ) )
cFLIP = ( NFkB )
BCL2 = ( NFkB )
BAX = ( ( CASP8 ) AND NOT ( BCL2 ) )
MPT = ( ( ROS ) AND NOT ( BCL2 ) )
DISC-TNF = ( TNFR AND ( ( ( FADD ) ) ) )
RIP1k = ( RIP1 )
apoptosome = ( ( Cyt_c AND ( ( ( ATP ) ) ) ) AND NOT ( XIAP ) )
survival = ( NFkB )
IKK = ( RIP1ub )
ATP = NOT ( ( MPT ) )
apoptosis = ( CASP3 )
MOMP = ( MPT ) OR ( BAX )
Cyt_c = ( MOMP )
CASP8 = ( ( DISC-TNF ) AND NOT ( cFLIP ) ) OR ( ( DISC-FAS ) AND NOT ( cFLIP ) ) OR ( ( CASP3 ) AND NOT ( cFLIP ) )
TNFR = ( TNF )
ROS = ( ( MPT ) AND NOT ( NFkB ) ) OR ( ( RIP1k ) AND NOT ( NFkB ) )
cIAP = ( ( NFkB ) AND NOT ( SMAC ) ) OR ( ( cIAP ) AND NOT ( SMAC ) )
SMAC = ( MOMP )
DISC-FAS = ( FASL AND ( ( ( FADD ) ) ) )
CASP3 = ( ( apoptosome ) AND NOT ( XIAP ) )
|
A Boolean Model of the Gene Regulatory Network
Underlying Mammalian Cortical Area Development
Clare E. Giacomantonio1, Geoffrey J. Goodhill1,2*
1 Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia, 2 School of Mathematics and Physics, The University of Queensland, St Lucia,
Queensland, Australia
Abstract
The cerebral cortex is divided into many functionally distinct areas. The emergence of these areas during neural
development is dependent on the expression patterns of several genes. Along the anterior-posterior axis, gradients of Fgf8,
Emx2, Pax6, Coup-tfi, and Sp8 play a particularly strong role in specifying areal identity. However, our understanding of the
regulatory interactions between these genes that lead to their confinement to particular spatial patterns is currently
qualitative and incomplete. We therefore used a computational model of the interactions between these five genes to
determine which interactions, and combinations of interactions, occur in networks that reproduce the anterior-posterior
expression patterns observed experimentally. The model treats expression levels as Boolean, reflecting the qualitative
nature of the expression data currently available. We simulated gene expression patterns created by all 1:68|107 possible
networks containing the five genes of interest. We found that only 0:1% of these networks were able to reproduce the
experimentally observed expression patterns. These networks all lacked certain interactions and combinations of inter-
actions including auto-regulation and inductive loops. Many higher order combinations of interactions also never appeared
in networks that satisfied our criteria for good performance. While there was remarkable diversity in the structure of the
networks that perform well, an analysis of the probability of each interaction gave an indication of which interactions are
most likely to be present in the gene network regulating cortical area development. We found that in general, repressive
interactions are much more likely than inductive ones, but that mutually repressive loops are not critical for correct network
functioning. Overall, our model illuminates the design principles of the gene network regulating cortical area development,
and makes novel predictions that can be tested experimentally.
Citation: Giacomantonio CE, Goodhill GJ (2010) A Boolean Model of the Gene Regulatory Network Underlying Mammalian Cortical Area Development. PLoS
Comput Biol 6(9): e1000936. doi:10.1371/journal.pcbi.1000936
Editor: Karl J. Friston, University College London, United Kingdom
Received March 25, 2010; Accepted August 17, 2010; Published September 16, 2010
Copyright: 2010 Giacomantonio, Goodhill. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which
permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work was supported by an Australian Postgraduate Award (CEG) and a Human Frontier Science Program grant RPG0029/2008-C. The funders had
no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: g.goodhill@uq.edu.au
Introduction
The mammalian cerebral cortex is a complex but extremely
precise structure. In adult, it is divided into several functionally
distinct areas characterised by different combinations of gene
expression, specialised cytoarchitecture and specific patterns of
input and output connections. But how does this functional
specification arise? There is strong evidence that both genetic and
activity-dependent mechanisms play a role in the development of
these specialised areas, a process also referred to as arealisation. A
genetic component is implicated by the spatial non-uniformity of
expression of some genes prior to thalamocortical innervation, as
well as the fact that altering expression of some genes early in
development changes area position in adult [for review see 1–8].
On the other hand, manipulating thalamocortical inputs, and
hence activity from the thalamus, can alter area size or respecify
area identity [for review see 1,4,8]. These results are accommo-
dated in a current working model of cortical arealisation as a
multi-stage process where initial broad spatial patterns of gene
expression provide a scaffold for differential thalamocortical
innervation [5]. Patterned activity on thalamocortical inputs then
drives more complex and spatially restricted gene expression
which, in turn, regulates further area specific differentiation. This
paper focuses on the earliest stage of arealisation: how patterns of
gene expression form early in cortical development.
Experiments have identified many genes expressed embryoni-
cally that are critical to the positioning of cortical areas in adult.
Although arealisation occurs in a two-dimensional field, most
experiments focus on anterior-posterior patterning and hence,
here we concentrate on patterning along this axis. From around
embryonic day 8 (E8) in mouse, the morphogen Fgf8 is expressed
at the anterior pole of the developing telencephalon (Figure 1A)
[2,3,5,7–11]. Immediately after Fgf8 expression is initiated in
mouse, four transcription factors (TFs), Emx2, Pax6, Coup-tfi and
Sp8 are expressed in gradients across the surface of the cortex
(Figure 1B) [2,3,5,8,11]. These four TFs are an appealing research
target because their complementary expression gradients could
provide a unique coordinate system for arealisation [5], equivalent
to ‘‘positional information’’ [12,13]. Altered expression of each of
Fgf8 and the four TFs shifts area positions in late embryonic stages
and in adult [14–29; but see also 30]. Furthermore, during
development, altered expression of each of these genes up- or
down-regulates expression of some other genes in the set along the
anterior-posterior axis (see Figure 2B for references). A large
PLoS Computational Biology | www.ploscompbiol.org
1
September 2010 | Volume 6 | Issue 9 | e1000936
cohort of experiments has given rise to a hypothesised network of
regulatory interactions between these five genes (Figure 2A).
However, only one of these interactions has been directly
demonstrated [24] and no analysis has been performed at the
systems level.
Interacting TFs are known to be able to form regulatory networks
that drive differential spatial development, fulfilling a role for which
morphogens are better known [31,32]. Feedback loops are the
crucial feature that enable the generation of spatial (and temporal)
patterns of expression of the genes in the network. Since TFs
regulate the expression of other genes, local differences in expression
of a set TFs are a powerful method of generating spatial patterns of
growth, differentiation and expression of guidance cues (and
therefore innervation), and developing more complex patterns of
gene expression. The arealisation genes form a regulatory network
with many feedback loops which is in principle capable of
generating spatial patterns. Establishing which interactions are
critical for correct arealisation is of great interest to the field, but
current experimental approaches are limited in their ability to
quickly assay the importance of each particular interaction.
Computational
modelling
of
gene
regulatory networks is
necessary because their complex behaviour is difficult to understand
intuitively. In addition, it offers several other benefits. Currently, the
many hypothesised interactions between arealisation genes are
represented as arrow diagrams like that seen in Figure 2A. Because
intuition tends to follow simple causal chains, the presence of many
feedback loops makes intuition about the overall behaviour of
complex systems unreliable [33–37]. Consequently, a more formal
description than an arrow diagram would test the current
conceptual model, and has the potential to give greater under-
standing and insight, as it has done for many other regulatory
networks
[for
review
see
33–36,38–44].
The
unambiguous
descriptions found in mathematical and computational models
offer the added benefit of making assumptions explicit and therefore
allowing greater scrutiny [45]. Computational experiments can also
be performed quickly and cheaply relative to laboratory experi-
ments and consequently can be useful for conducting thought
experiments which can then be tested experimentally [45,46]. In
this way, computational modelling and experiments can spur each
other on so that both are ‘‘improved in a synergistic manner’’ [36].
Here, we use the Boolean logical approach to model the arealisation
regulatory network. In this approach, variables representing genes and
proteins can take only two values, zero or one, representing gene and
protein activity being below or above some threshold for an effect.
While continuous models are more realistic, they have many free
parameters which are hard to constrain from experimental data, and
offer a formidable computational challenge to investigate systemati-
cally. In contrast, Boolean models can be used when only qualitative
expression and interaction data are available, as is the case for
arealisation. In Boolean models, at each point in time, the state of a
variable depends on the state of its regulators at the previous time step.
A set of logic equations capture the regulatory relationships between
Figure 1. Gene expression in the developing neocortex. (A) The anterior neural ridge or commissural plate (blue) is a patterning centre in the
developing forebrain that secretes the morphogen Fgf8. Since the protein is secreted, it is hypothesised that it diffuses to form a gradient [5]. The
directions A, P, D, V, M and L indicate anterior, posterior, dorsal, ventral, medial and lateral respectively. (B) These four transcription factors are
expressed in spatial mRNA and protein gradients across the developing forebrain. Many other genes with spatial patterns of expression have also
been identified [for review see 8]. (C) A schematic of the desired steady state expression levels in the anterior and posterior compartments in the
discretised Boolean model. A is adapted from Figure 1A in [4] and Figure 1 in [5], B is adapted from Figure 6A in [5].
doi:10.1371/journal.pcbi.1000936.g001
Author Summary
Understanding the development of the brain is an
important challenge. Progress on this problem will give
insight into how the brain works and what can go wrong
to cause developmental disorders like autism and learning
disability. This paper examines the development of the
outer part of the mammalian brain, the cerebral cortex.
This part of the brain contains different areas with
specialised functions. Over the past decade, several genes
have been identified that play a major role in the
development of cortical areas. During development, these
genes are expressed in different patterns across the
surface of the cortex. Experiments have shown that these
genes interact with each other so that they each regulate
how much other genes in the group are expressed.
However, the experimental data are consistent with many
different regulatory networks. In this study, we use a
computational
model
to
systematically
screen
many
possible networks. This allows us to predict which
regulatory interactions between these genes are important
for the patterns of gene expression in the cortex to
develop correctly.
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
2
September 2010 | Volume 6 | Issue 9 | e1000936
variables and dictate how the system evolves in time. The Boolean
idealisation greatly reduces the number of free parameters while still
capturing network dynamics and producing biologically pertinent
predictions and insights [43,45,47]. In our model, we use only two
spatial compartments, one representing the anterior pole and another
representing the posterior pole. The anterior and posterior expression
levels after Boolean discretisation are shown in Figure 1C. More than
two expression levels and more than two spatial compartments would
be more realistic, but would result in an explosion in the number of
parameters currently unconstrained by experimental data. Having
only two expression levels and only two compartments allows us to
systematically screen a large number of networks, which would be
impossible in a more complex model.
In this paper, we simulate the dynamics of all possible networks
created by different combinations in interactions between Fgf8,
Emx2, Pax6, Coup-tfi and Sp8, and show that only 0:1% of these
networks are able to reproduce the expression patterns observed
experimentally. From this analysis, we identify structural elements
common to the best performing networks, as well as elements that
never appear in the networks that perform well. These results
reveal important logical principles underlying the cortical area-
lisation gene network, and suggest potential directions for future
experimental investigations.
Results
Simulation of the dynamics of 224 possible networks
revealed networks that reliably reproduced the
experimentally observed expression gradients
Experimental evidence indicates that Fgf8, Emx2, Pax6, Coup-tfi
and Sp8 regulate each other’s expression, but the actual structure
of the network is highly unconstrained by experimental data.
Figure 2. A network created by interactions between the five genes of interest as suggested by experiments. (A) Arrows (?) indicate
inductive or activating interactions, flat bars (a) indicate repressive interactions. Text in italics signifies genes while upright text signifies proteins.
Only the activation of Fgf8 by Sp8 (Sp8?Fgf8) has been directly demonstrated [24]. Other interactions have generally been inferred based on altered
expression patterns in mutants and therefore might be indirect. For example, the activation of Emx2 by Coup-tfi might be due to Coup-tfi repressing
Pax6 which in turn represses Emx2. This panel is adapted from Figure 6B in [5]. (B) References for each of the interactions in panel A. (C) The Boolean
logic equations for the network in panel A. F, E, P, C and S are the logical variables representing the genes Fgf8, Emx2, Pax6, Coup-tfi and Sp8 and F, E,
P, C and S are the logical variables representing the respective proteins. For a gene to be turned on at time tz1, its inductive regulators must be
present and its repressive regulators absent at time t.
doi:10.1371/journal.pcbi.1000936.g002
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
3
September 2010 | Volume 6 | Issue 9 | e1000936
Hence, we performed a systematic screen of the different possible
networks and then looked for common structural features in the
networks that perform poorly and well.
We analysed the dynamics of all networks created by different
combinations of 24 possible interactions between these five genes
and their respective proteins. In each network, Sp8?Fgf8 was
fixed since this has been directly demonstrated [24]. We also did
not consider positive interactions between species with opposing
expression gradients, or negative interactions between species with
the same gradient. For example, Emx2aPax6 and Fgf8?Pax6
were possible interactions, but Emx2?Pax6 and Fgf8aPax6 were
not. The 24 variable interactions generated 224~1:68|107
possible networks. The structure of each network was transformed
into a set of Boolean logic functions as described in the Methods.
We identified networks that proceeded from the state at the
anterior pole at E8 to the state at around E10.5, as well as from the
state at the posterior pole at E8 to the state at around E10.5. At
E8, of our genes of interest, only Fgf8 is active due to mechanisms
external to the network we are modelling [5,24,26], and only at
the anterior pole. Hence, in the Boolean model with binary
variables, Fgf8 gene and protein started in the active (‘1’) state in
the anterior compartment and inactive (‘0’) state in the posterior
compartment, while the other genes and proteins started in the
inactive state in both compartments. By E10.5, the expression
patterns seen in Figure 1 are present. That is, at the anterior pole,
Fgf8, Pax6 and Sp8 genes are active, while Emx2 and Coup-tfi are
inactive; at the posterior pole, Emx2 and Coup-tfi are active, while
Fgf8, Pax6 and Sp8 are inactive.
When the Boolean update functions describing a network were
applied stochastically, many networks reached multiple steady states
with fixed probabilities. In these cases, we calculated the average
gene and protein levels, weighted by the probability of ending in a
particular state, and thus each Boolean variable could be between 0
and 1. We say that a network reliably reaches a desired steady state
if it does so with a greater than 50% probability. From this, it follows
that networks that reliably reach both the anterior and posterior
steady states from the respective starting states have differences in
activity between the anterior and posterior poles than span 0.5, as in
Figure 1C. We define these networks as good.
A previously hypothesised regulatory network does not
satisfy our criteria for reproducing the experimental
observations
To give a specific example, we present the dynamics of a
regulatory network previously hypothesised based on experimental
observations [5,8], seen in Figure 2A. The network was converted
into the set of Boolean logic equations described in Figure 2C. We
found that this network had a 100% chance of following the
desired trajectory from the posterior starting state to the posterior
steady state. In constrast, it had only a 38% chance of following
the desired anterior trajectory from the anterior starting state to
the anterior steady state. This poor anterior performance arises
because of Fgf8 auto-induction and the Fgf8/Sp8 inductive loop,
as we will explain later in more detail. While this network
produced the correct activity gradients overall, as seen in
Figure 3A, it does not satisfy our criteria for doing so reliably
Figure 3. Performance of a previously hypothesised network, and all possible networks. (A) The average expression in the anterior and
posterior compartments in the experimentally hypothesised network in Figure 2. This network does not satisfy our criteria for reliable performance
because the gradients do not span 0.5. (B) Each network followed the desired anterior state trajectory and the desired posterior state trajectory with
a fixed probability plotted on the two axes of this graph. We defined good networks as those with a greater than 50% chance of following both the
desired anterior trajectory of states as well as the desired posterior trajectory of states. These networks lie in the upper, right quandrant of this graph
(blue plusses). All other networks (black crosses) did not satisfy our criteria for reliably reproducing the experimentally observed patterns of gene
expression. The point corresponding to the experimentally hypothesised network in Figure 2 is coloured green. The red plus corresponds to the two
best performing networks in Figure 7B and C. The black contour lines are lines of constant network performance.
doi:10.1371/journal.pcbi.1000936.g003
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
4
September 2010 | Volume 6 | Issue 9 | e1000936
because the gradients do not span 0.5; the anterior levels of Fgf8,
Pax6 and Sp8 are too low (v0:5), while the anterior levels of Emx2
and Coup-tfi are too high (w0:5). The fact that this network did not
reach our criteria for reproducing the experimental observations,
even though all interactions have been observed indirectly, shows
clearly that intuitions about the dynamics of regulatory networks
with feedbacks can be unreliable.
Only a small percentage of networks satisfied our criteria
for reproducing the experimental observations
Of all possible networks, we found that 0.1% of networks
(1:00|104) had a greater than 50% chance of proceeding from
the anterior starting state to the anterior steady state, as well as a
greater than 50% of proceeding from the posterior starting state to
the posterior steady state. In a plot of the probability of following
the desired anterior trajectory through state space against the
probability of following the desired posterior trajectory, these good
networks lie in the upper right quadrant (Figure 3B).
To assess the similarity of the structures of the good networks,
we calculated the average distance from each of the good networks
to the best performing network and compared this to the average
distance from all networks (Figure 4). The distance is defined as the
number of different interactions [48]. The good networks differ to
the best network by an average of 7:3+2:1 interactions, while all
possible networks differ by an average of 12:0+2:4 interactions.
This indicates that the structures of the good networks are
restricted in the space of all possible networks. We then set out to
understand which network interactions characterised the good and
bad networks.
There were many combinations of interactions that did
not appear in networks that performed well
Careful examination of the interactions present and absent in
the good networks allowed us to identify several combinations of
interactions that were never present in good networks. Networks
containing nodes with no regulators obviously performed poorly.
Figure 5A shows their position on the plot of probability of
following the desired anterior trajectory though state space against
the desired posterior trajectory. In a similar vein, networks where
Fgf8 was not upstream of at least one of the four TFs also
performed poorly (Figure 5B), because the starting states of the two
compartments only differed in Fgf8 activity. In addition, networks
with auto-inductive interactions all performed poorly (Figure 5C).
This occurs because any node with auto-induction is either locked
into its initial state or becomes inactive if it has other regulatory
requirements that are not satisfied. Consequently, the desired
trajectories cannot occur with a greater than 50% probability in
both compartments. By similar reasoning, nodes with inductive
loops also performed poorly (Figure 5D), as do networks with
isolated repressive loops (Figure 5E).
We also identified several higher order combinations of
interactions that rarely appeared in networks that could produce
the average expression gradients observed experimentally. For
these higher order combinations, we could not deduce an intuitive
explanation for why they caused networks to perform poorly.
Some of these combinations are listed in Table S1. Removal of
networks containing these interactions further narrowed the space
of possible networks as seen in Figure 6. In total, the criteria
outlined so far reject 99.96% of the networks investigated, leaving
6980 networks.
Some interactions were more likely than others to occur
in the networks that performed well
By analysing the remaining networks, we identified certain
interactions that were more likely than others. Of the remaining
networks, 84% (5849 of 6980), satisfied our criteria for reliably
following the desired trajectories to produce the average expression
gradients observed experimentally. Surprisingly, among these good
networks, no single interaction was universally present or absent,
except those already identified as deterimental (Table 1, third
column). In fact, among the remaining, good networks, all
interactions occurred at about the frequency expected from all the
remaining networks (Table 1, third column compared to second
column). In general, the repressive interactions were more likely
than inductive ones. The interactions Fgf8aEmx2 and Fgf8aCoup-tfi
were the most likely interactions, occuring in 80% of all remaining
networks that performed well. Next, Emx2aSp8 and Coup-tfiaSp8
occurred in 66% of good networks. The interactions Pax6aEmx2
and Pax6aCoup-tfi occurred in 55% of all good networks, while
Emx2aPax6 and Coup-tfiaPax6 occurred in 54% of all good
networks.
Though many different networks performed well, we now
discuss the best performing networks as an illustrative example.
The two best performing networks both followed the desired
anterior trajectory 74% of the time and the desired posterior
trajectory 74% of the time. They are marked in red in Figure 3B
and reliably produced the average expression gradients observed
experimentally (Figure 7A cf. Figure 1C). Figure 7B and C show
the structures of these two networks. Note that the six most likely
interactions from the third column of Table 1 are present in these
networks, as well as several less common interactions. However,
many networks with similar structures also produced the correct
average expression gradients, while some with quite different
structures did too. Thus, although the networks that reproduced
the experimentally observed expression gradients were constrained
in structure compared to all possible structures, there was still a
remarkable diversity in these networks.
In general, repressive interactions were more prevalent in the
networks that performed well than inductive interactions. This is
evident in the probabilities of each interaction being present
(Table 1, third column), as well as a set of networks that performed
Figure 4. Distribution of the structural difference between the
best network and the good networks, as well as the best
network and all networks. The distance between two networks is
the number of interactions differing between them. The good networks
are constrained in their structure so that there is less difference
between them and the best network than between all networks and
the best network.
doi:10.1371/journal.pcbi.1000936.g004
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
5
September 2010 | Volume 6 | Issue 9 | e1000936
similarly to the best performing network, that are illustrated in
Figure 8A. In these 64 networks, the six most common interactions
in the good networks were all required to be present. All other
repressive interactions, which created reciprocal repressive loops,
could be present or absent without greatly affecting network
performance. The only inductive interaction appearing in this set
of networks was Fgf8?Pax6, and it was present in all 64 networks.
All inductive interactions between the four TFs were required to
be absent along with Pax6?Fgf8, all auto-inductive loops and
Fgf8?Sp8 which created an inductive loop.
Discussion
The roles of different genes
Current experimental evidence indicates that the gene network
that regulates cortical area development has multiple feedback
pathways and consequently, it is difficult to understand intuitively.
Using a Boolean logic model, we simulated many different possible
networks and identified many structural requirements on the
networks to ensure good performance.
Our analysis suggests differing roles for the different genes in the
network. We show that Fgf8 expression at the anterior pole, a
putative cortical patterning centre, may be sufficient to drive the
correct spatial patterning of the transcription factors Emx2, Pax6,
Coup-tfi and Sp8, if simple interactions between these transcription
factors exist. This is an example of how a transient signal, in this
case Fgf8 expression initiated by external regulators, can be
converted into a durable change in the developing brain [49].
In our simplified model, Emx2 and Coup-tfi, which are both
expressed in high posterior–low anterior gradients, play the same
role in the network. This means that if Emx2 and Coup-tfi are
swapped in any network, the dynamics of the network don’t
change. This is evident in the higher order interactions that rarely
appear in good networks, listed in Table S1, as well as the two best
performing networks in Figure 7B. In reality, Coup-tfi has a sharper
anterior-posterior expression gradient than Emx2 and the two TFs
Figure 5. Some combinations of interactions that never appear in good networks. Each panel shows the probability of following the
desired anterior state trajectory against the probability of following the desired posterior state trajectory for all 224 networks that we considered. In
each panel, we highlight in red networks that contain a particular combination of interactions. All other bad networks are marked with black crosses,
all other good networks are marked with blue pluses. (A) In red are networks containing nodes with no regulators. These entered the anterior steady
state or posterior steady state but not both. (B) In red are networks with Fgf8 only downstream of the four TFs (Fgf8aEmx2, Fgf8?Pax6, Fgf8aCoup-tfi
and Fgf8?Sp8 all absent). Because the only difference in the starting state between the two compartments was Fgf8 activity, these networks could
not enter both the anterior and posterior steady states with w50% probability. (C) Marked in red are networks with auto-induction. Networks with
Emx2, Pax6, Coup-tfi or Sp8 auto-induction entered the anterior steady state or the posterior steady state but not both. Networks with Fgf8 auto-
induction could reliably enter the posterior steady state but not the anterior steady state. To enter the anterior steady state, they required Sp8 to
become and remain active before the state of Fgf8 was updated. Because nodes were updated asynchronously in a random order, this could not
occur with w50% probability. (D) In red are networks containing inductive loops. These also could not enter the anterior steady state with w50%
probability by similar reasoning to C. (E) In red are networks containing isolated repressive loops (that is, X repressing Y was the only regulation of Y
and Y also repressed X). These also could not reproduce the average gradients observed experimentally.
doi:10.1371/journal.pcbi.1000936.g005
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
6
September 2010 | Volume 6 | Issue 9 | e1000936
are expressed in opposing gradients along the medial-lateral axis.
Experiments suggest that Emx2 promotes posterior area identity
while Coup-tfi represses anterior area identity [6]. Therefore, we
expect that they are not redundant as our model suggests, but play
different roles through differing downstream targets.
Interactions we predict are likely
Our screen of possible networks identified interactions that we
predict are more likely to be present in the arealisation regulatory
network than others, and are therefore good experimental targets
for further study. In general, we predict that repressive interactions
are particularly important in this network. This is consistent with
data showing that repressive cascades are important for spatial
differentiation in other systems [50,51]. The interactions we predict
are most likely include several interactions that have previously been
hypothesised based on experiments. Our analysis predicts that
Fgf8aEmx2 and Fgf8aCoup-tfi are the most likely direct interactions,
consistent with many previous suggestions [20,21,27,52–54]. Since
Sp8 induces Fgf8, repression of Sp8 by Emx2 has been proposed as a
mechanism by which Fgf8 expression can be contained to the
anterior pole [24]. Our analysis predicts that repression of Sp8 by
Emx2 or Coup-tfi, or both, is quite likely. Currently, possible
repression of Sp8 by Coup-tfi has not been discussed in the
experimental literature. Reciprocal repression between Emx2 and
Pax6 has been frequently discussed as potential regulatory
interaction [5,7,11,23,55,56; but see also 2]. Our analysis predicts
that these interactions are approximately equally likely. However, it
also predicts that reciprocal repression loops in general are not
critical for correct functioning of the network.
Interactions we predict are unlikely
Our screen of networks also predicts several single interactions
and many combinations of interactions that are unlikely to occur in
the arealisation regulatory network since they usually lead to poor
performance. The lack of an intuitive explanation of why some of
the combinations of interactions degrade network performance
demonstrates the complexity of the network dynamics, and why
computational modelling of these networks gives insights not
available through intuition. Several of the interactions that we
predict are unlikely have previously been hypothesised based on
experiments. In particular, Fgf8?Sp8 has been proposed by
Sahara et al. [24] but our simulations predict that this interaction
creates an inductive loop which is detrimental to network
performance. The experimental evidence for this interaction is
that ectopic expression of Fgf8 in the telencephalon by in utero
electroporation at E11.5 induced ectopic expression of Sp8 at
E13.5 [24]. However, the target tissue contained an active
regulatory network that could have indirectly initiated expression
of Sp8 after perturbation by ectopic Fgf8. Direct auto-induction of
any of the five genes prevented networks from being able to
recreate the experimental expression patterns. Auto-induction of
Fgf8 has previously been hypothesised based on experiments
implanting Fgf8-coated beads in the chick midbrain [57], limbs
[58] and telencephalon [52], but our model predicts the resulting
induction of ectopic Fgf8 is unlikely to be direct. It could be
occurring
indirectly
through
an
active
regulatory
network
perturbed by ectopic Fgf8. For example, in the forebrain, if
Emx2 does limit the region of Fgf8 expression by repressing Sp8
inducing Fgf8 (Emx2aSp8?Fgf8, [24]) and Fgf8 represses Emx2,
then ectopic Fgf8 protein could induce the transcription of ectopic
Fgf8 mRNA. A more definitive test of Fgf8 auto-induction would
require to addition of ectopic Fgf8 the absence of Sp8 or Emx2.
Changes in Fgf8 expression would need to be examined after
E12.5 because Fgf8 expression in the forebrain appears to be
initiated by regulators outside the network studied here and only
maintained by Sp8 [26].
Relation to other modelling work
To date, we are only aware of one paper modelling cortical
arealisation [59]. The model starts with expression gradients of
Figure 6. Some higher order combinations of interactions rarely appear in good networks. Each panel shows the probability of following
the desired anterior state trajectory against the probability of following the desired posterior state trajectory for all 224 networks that we considered.
In each panel, we highlight in red networks that contain particular combinations of interactions. All other bad networks are marked with black
crosses, all other good networks are marked with blue pluses. (A) In red are networks containing any of the combinations of three interactions listed
in Table S1. (B) In red are networks containing any of the combinations of four interactions that we found caused networks to perform badly.
doi:10.1371/journal.pcbi.1000936.g006
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
7
September 2010 | Volume 6 | Issue 9 | e1000936
Fgf8, Emx2 and Pax6, which are maintained by regulation of each
other. It then goes on to simulate the formation of area-specific
thalamocortical connections. In contrast, this paper focuses on
modelling pattern generation by the gene regulatory network, and
at present does not consider the later process of thalamocortical
innervation where less data are available to constrain models.
This paper draws on the ideas used in other Boolean modelling
papers in different systems, but a systematic analysis of possible
regulatory networks is novel. Although algorithms exist for reverse
engineering the Boolean expressions and hence the structure of
regulatory networks [60,61], they require data on the time course
of expression levels. For example, Laubenbacher and Stigler [61]
tested a reverse engineering algorithm by reconstructing a well-
characterised network. They showed that their algorithm only
worked well when it used time series data from mutant animals, as
well as wild type time series. Currently, these data are unavailable
for the system we have investigated for either wild type or mutant
animals.
More recently, Wittmann et al. [62] used Boolean modelling to
infer regulatory relationships governing the spatial patterning of
genes at the midbrain-hindbrain isthmus. They were able to use a
spatial, rather than temporal pattern to infer minimal Boolean
equations
using
reverse
engineering
strategies
from
digital
electronic engineering. Compared to the gene expression patterns
at the isthmus however, the arealisation expression patterns are
much simpler and consequently do not provide us with many
constraints for the reverse engineering algorithm. In any case, for
more complex modelling Wittmann et al. added additional
interactions hypothesised in the literature. In contrast, our
simulation of an experimentally hypothesised network gave a
negative result, which led to our systematic screening all possible
networks. Our goal was to explore the space of possible networks
rather than identify one individual network that could produce the
desired results as Wittmann et al. did, when many other sufficient
networks likely exist.
Albert and Othmer [63] explored a single well-characterised
network (the Drosophila segment polarity network) in great detail.
Using Boolean analysis, they were able to reproduce mutants and
predict novel mutants. Unfortunately, mutants in the arealisation
genes exhibit a phenotype of shifted expression gradients of the
other genes (see Introduction). These results cannot be reproduced
in the two compartment, two level model used in this paper (see
Methods, Spatial Compartments for more detail).
An extended model with additional spatial compartments and
expression levels, or continuous expression levels would be able to
incorporate the mutant data. However, these types of models have
many more parameters that cannot be constrained by the
qualitative experimental data available in this case. Any systematic
exploration or optimisation of parameter space for the large
number of possible networks we simulate in this paper would be
computationally impossible. For example, an ordinary differential
equation model using Michaelis-Menten kinetics has two param-
eters per interaction (the Hill coefficient and the Michaelis
constant), as well as a degradation rate and a constitutive activity
rate for each species [35,43], none of which are constrained by
experimental data.
Communication between cells
In this paper, we have not considered any communication
between cells since we model only two compartments at the
anterior and posterior poles. However, communication between
cells may occur and may be useful. Although we find that many
networks
can
produce
the
experimentally
observed
average
expression patterns, we find that in most cases, each network
has more than one accessible steady state from each of the starting
states. We speculate that this may be resolved by cell-cell signalling
of some kind, most likely by Fgf8, which is known to be a secreted
molecule. Such signalling could lock the regulatory networks of
nearby communicating cells into the same state. Fgf8 movement
by diffusion or some other kind of transport might also generate
the smooth gradients of the TFs. An investigation of the effects of
Fgf8 diffusion would require a more complex model with more
than two discrete expression levels and more than two compart-
ments.
Conclusions
Overall, our exploration of the dynamical consequences of
different structures of the network consistent with experimental
data predicts constraints on the structure of the real network. The
Boolean approach we used is well suited to the qualitative data
currently available, and permitted us to screen a large number of
networks. Our results may be used as a starting point for future
more realistic models of the gene networks regulating cortical
arealisation because the narrowed pool of possible networks may
Table 1. Probability of interactions being present in all
remaining networks, compared to remaining networks that
perform well.
Interaction
P(present) in
all remaining
networks (%)
P(present) in all
remaining good
networks (%)
FaE
80
80
FaC
80
80
EaS
66
66
CaS
66
66
PaE
55
55
PaC
55
55
EaP
54
54
CaP
54
54
S?P
53
51
F?P
44
46
EaF
42
43
CaF
42
43
SaE
36
39
SaC
36
39
C?E
29
31
E?C
29
31
P?F
23
18
P?S
13
15
F?F
0
0
E?E
0
0
P?P
0
0
C?C
0
0
S?S
0
0
F?S
0
0
All interactions in the good networks occur at about the expected frequency,
which means that it is difficult to identify any further combinations of
interactions that ensured networks performed badly or well. Despite this, we
can still use the probabilities of individual interactions among the remaining
good networks as an indicator of which interactions are likely and unlikely to
occur in the gene network regulating cortical arealisation.
doi:10.1371/journal.pcbi.1000936.t001
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
8
September 2010 | Volume 6 | Issue 9 | e1000936
make it feasible to investigate parameter space systematically in a
more realistic model with many more free parameters. From an
experimental perspective, data on the time course of expression
levels at different spatial locations, or even accurate relative
protein levels would provide useful constraints to future models.
We show here though that even a simple Boolean model reveals
logical principals underlying the genetic regulation of cortical
arealisation, and may be used to guide future experiments.
Figure 7. Best performing networks. (A) In the best performing networks, the average activity of the genes and proteins of interest in the
anterior and posterior compartments formed gradients in the same direction as those observed in mouse (cf Figure 1C). (B) The structure of the two
best networks. The purple boxes with names in italics represent genes and the blue ellipses with names in upright text represent proteins. Each of the
gene?protein interactions has been condensed into a green box to simplify the diagram and avoid intersecting edges. Each edge between the
rounded green boxes indicates how the protein in the source box regulates the gene in the target box. The two best networks performed equally
well. However, some other networks with quite different structures also performed nearly as well.
doi:10.1371/journal.pcbi.1000936.g007
Figure 8. A selection of networks that produced the correct average expression gradients and have common structural elements.
(A) The structure of the networks. The purple boxes with names in italics represent genes and the blue ellipses with names in upright text represent
proteins. Each of the gene?protein interactions has been condensed into a green box to simplify the diagram and avoid intersecting edges. Each
edge between the rounded green boxes indicates how the protein in the source box regulates the gene in the target box. The solid lines indicate
interactions that must be present while the dashed lines indicate interactions that can be present or absent. The 6 dashed interactions means that
this diagram represents 26~64 different networks. (B) The performance of these 64 networks (red pluses) on a plot of probability of following the
desired anterior state trajectory against the probability of following the desired posterior state trajectory. All other good networks are marked with
blue pluses, all bad networks with black crosses.
doi:10.1371/journal.pcbi.1000936.g008
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
9
September 2010 | Volume 6 | Issue 9 | e1000936
Materials and Methods
Networks simulated
We examined the dynamics of all possible networks created by the
five genes and five proteins of interest in anterior-posterior patterning
of cortical areas: Fgf8, Emx2, Pax6, Coup-tfi and Sp8 and their
respective proteins. The induction of Fgf8 by Sp8 has been directly
demonstrated [24], and therefore, this interaction was fixed in all the
simulated networks. Genes were also always fixed to induce their
corresponding protein. To narrow the number of networks
considered, other interactions were either inductive (?) or repressive
(a), depending on the anterior-posterior expression patterns observed
experimentally (shown in Figure 1). For example, because Emx2 and
Pax6 are expressed in counter gradients, we considered the
interactions Emx2aPax6 and Pax6aEmx2 but not the interactions
Emx2?Pax6 and Pax6?Emx2. This gave 24 possible interactions,
summarised in Table 2, which have not been directly demonstrated.
Hence, we considered all 224~1:68|107 networks formed by
different combinations of the possible interactions.
Converting a network into Boolean logic functions
Each network was turned into a set of Boolean logic functions
using the logical operators AND and NOT. Repressive interac-
tions were incorporated with a negation (NOT operator). We
assumed that if a gene has multiple regulators, all regulatory
conditions must be met, and so we combined their action with a
logical conjunction (AND operator). For example, the network in
Figure 2A was transformed into the set of Boolean functions in
Figure 2C. According to these equations, the state of a gene or
protein at time tz1 is governed by the state of its regulators at
time t. A protein will only be active if its corresponding gene is
active at the previous time step, and a gene will only be active if
the transcriptional activators of that gene are active at the previous
time step and the inhibitors are inactive.
Implicit in these functions are several assumptions [63]: (1) if the
regulatory requirements for transcription or translation to occur
are satisfied, then the mRNA or protein is synthesised in one time
step, (2) mRNA decays within one time step if the necessary
regulatory requirements do not continue to be satisfied, and (3)
active protein decays within one time step. Albert and Othmer
[63] tried relaxing these assumptions and found that it did not
change the steady states. We did not consider the OR logical
operator, which corresponds to the situation where only one
regulatory condition (or a subset of conditions) must be satisfied to
set a gene to the active state, or other logical operators. While it
would obviously be possible to relax these assumptions, this would
cause a large increase in the complexity of the model and a
combinatorial
explosion
in
the
number
of
parameters
to
investigate, making it harder to analyse and derive conclusions
from the model.
Spatial compartments
Since we were interested in anterior-posterior patterning, it was
necessary to have a spatial dimension in the model. This was
incorporated by considering two compartments, one anterior, one
posterior. The regulatory networks, and therefore logic functions,
operating in the two compartments were the same. The difference
between the two compartments was their initial conditions
(outlined later). There was no signalling between compartments.
There are several reasons why signalling between compartments
was not incorporated into the model. Firstly, there is currently no
experimental evidence for long range communication between
cells via our molecules of interest. As discussed in the Introduction,
Fgf8 is a secreted protein, and it is hypothesised to diffuse, but only
its mRNA expression has been characterised. Even if it does
diffuse, it is unlikely to be present at high concentration at the
posterior pole, represented in our model by the posterior
compartment. Gradients of TF mRNA (and presumably protein)
must form by some mechanism other than diffusion and here we
assume the TFs act on each other independently in each
compartment.
Given the lack of signalling between compartments, and the fact
that in Boolean models, each gene and protein can only have the
state ‘0’ or ‘1’, two compartments with different initial conditions
were sufficient to completely explore the system. Additional
compartments between the anterior and posterior extremes would
have to start with the same initial conditions as either the anterior
or posterior compartment. Without communication between
Table 2. Summary of all the considered interactions.
Regulator
Target gene or protein
Fgf8
Fgf8
Emx2
Emx2
Pax6
Pax6
Coup-tfi
Coup-tfi
Sp8
Sp8
Fgf8
+
Fgf8
z
{
z
{
z
Emx2
+
Emx2
{
z
{
z
{
Pax6
+
Pax6
z
{
z
{
z
Coup-tfi
+
Coup-tfi
{
z
{
z
{
Sp8
+
Sp8
+
{
z
{
z
All possible combinations of these interactions form the space of networks whose dynamics were simulated. Text in italics signifies genes while upright text signifies
proteins. A ‘z’ indicates an inductive interaction while a ‘{’ indicates a repressive interaction. The table is sparse because we assume that proteins can’t regulate
proteins and a gene can only regulate its corresponding protein. The circled interactions (+) were present in every network because these have been directly
demonstrated by experiments. These include each gene producing its respective protein and Sp8 activating Fgf8. The other 24 interactions are possible but have not
been directly demonstrated. We simulated the dynamics of the 224 networks formed by all combinations of the possible interactions.
doi:10.1371/journal.pcbi.1000936.t002
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
10
September 2010 | Volume 6 | Issue 9 | e1000936
compartments, these hypothetical additional interior compart-
ments would follow the same dynamics as an exterior compart-
ment with the same starting state. Hence, they would not provide
any extra information to constrain the structures of the arealisation
network.
A consequence of this two-level, two-compartment model is that
we cannot simulate the mutant phenotypes of shifting expression
gradients (see Figure 2B for references). In the current model, an
expression gradient is represented by a protein in the active state in
one compartment and in the inactive state in the other
compartment. The only other possible expression patterns are
active–active or inactive–inactive, which are not shifted gradients.
Initial states and desired steady states of each
compartment
The difference between the two compartments was their initial
state, and we were interested in which steady state they each ended
up in given the different initial states. We describe the state of the
system with a ten-tuple of 1’s and 0’s representing the state of the
network nodes [Fgf8, Fgf8, Emx2, Emx2, Pax6, Pax6, Coup-tfi, Coup-
tfi, Sp8, Sp8]. For example, the state [1,1,0,0,0,0,0,0,0,0] denotes
Fgf8 gene and protein are active, while all other genes and proteins
are inactive. This corresponds to the starting state in the anterior
compartment at around E8, where Fgf8 expression is thought to be
initiated via a mechanism external to the regulatory network we are
modelling [5,24,26]. We assume that the expression of the other
four genes is controlled by the regulatory network we are modelling.
In the posterior compartment, we assume that the expression of all
five genes is controlled by the modelled regulatory network and so
this compartment starts in the state [0,0,0,0,0,0,0,0,0,0]. The
Boolean versions of the desired anterior and posterior steady states
are seen in Figure 1C and are given in tuple notation as
[1,1,0,0,1,1,0,0,1,1] for the anterior compartment and [0,0,1,
1,0,0,1,1,0,0] for the posterior compartment. We were interested
in networks which flowed from the anterior starting state
[1,1,0,0,0,0,0,0,0,0] to the anterior steady state [1,1,0,0,1,1,0,0,
1,1], as well as from the posterior starting state [0,0,0,0,0,0,0,0,0,0]
to the posterior steady state [0,0,1,1,0,0,1,1,0,0].
Creating and analysing state tables
The binary tuple representation of states emphasises the fact
that Boolean networks are finite state machines whose steady states
can be readily determined. Initially, we determined the steady
states of each network by creating a state table for each network.
This is a list of all possible states of the network ( [0,0,0,0,0,0,0,0,-
0,0], [0,0,0,0,0,0,0,0,0,1],…, [1,1,1,1,1,1,1,1,1,1]) and the corre-
sponding next state when the Boolean rules were applied. Steady
states were those that did not change under the Boolean rules.
Unfortunately however, this analysis could not reveal which
networks proceeded along the desired trajectories through state
space. Trajectories can be traced in state tables, but such
trajectories assume that all nodes update synchronously so that
trajectories are deterministic. Synchronous updating has been used
previously in Boolean modelling [63] but synchronous trajectories
frequently end up in artefactual cyclic attractors [35,49,64]. In
reality, it is highly unlikely that multiple species in a network would
change their state at exactly the same time. Rather, these systems
are stochastic, with nodes updated asynchronously, and this has
consequences for the dynamics of the system.
Simulating the networks with asynchronous updating
Assuming fully asynchronous updating of nodes enabled the use
of the Markov chain formalism to describe and analyse the
network dynamics [65]. The transition matrix T of a Markov
chain contains the probability of transition from each state to other
states in state space. We used the deterministic state table
described above to calculate the transition matrix, T, of each
network, assuming that each individual node changed state with
equal probability. For example, a deterministic transition from
[1,1,0,0,0,0,0,0,0,0] to [1,1,0,0,1,1,0,0,0,0] translated to a sto-
chastic transition to state [1,1,0,0,1,0,0,0,0,0] or [1,1,0,0,0,1,0,
0,0,0], each with a 50% probability. We found that most networks
formed reducible Markov chains, with more than one steady state,
each a part of a closed class of states. In general, the anterior and
posterior starting states were transient states that could end up at
more than one steady state. The probability of ending up at
different steady states from a transient state could be calculated
analytically [65] or by performing the simple computation:
s(n)~Tns(0)
ð1Þ
where s(n) is the distribution of states of the system at time step n.
Note that s is different to the state tuple notation used so far.
Instead, it is a column vector of length 2No: nodes~210. The
probability of being in state [0,0,0,0,0,0,0,0,0,0] is given by
element s1, the probability of being in state [0,0,0,0,0,0,0,0,0,1] is
given by element s2, and so on. The two compartments in our
model each started in a single state, not a distribution of states.
Hence, the anterior starting state [1,1,0,0,0,0,0,0,0,0] correspond-
ed to an s-vector with a probability of one at element s769 and zero
probability elsewhere, and the posterior starting state [0,0,0,0,
0,0,0,0,0,0] corresponded to an s-vector with a probability of one
in element s1 and zero probability elsewhere. The element si(n)
gives the probability of finding the system in state i at time step n.
Since our networks always ended up in a distribution of steady
states, if n was large enough, the computation of Equation 1
determined the probability of ending up at different steady states
from the starting state s(0). In our analysis, since we knew the
steady states of each Markov chain from the state tables, we
iteratively calculated s(n)~Ts(n{1) until there was a 99.99%
chance of being in the steady states.
In many cases, there was a distribution of steady states. As each
compartment represented many cells, the steady state probability
distribution could be interpreted as the distribution of states across
an inhomogeneous cell population [66]. Hence, we calculated the
average amount of each species in a compartment as the sum of
steady states of each compartment weighted by the probability of
entering that steady state.
Analysing the similarity between different groups of
networks
We quantified the structural difference between two networks as
the number of interactions differing between them. We refer to
this as the distance between networks because if the network
structure is notated as a vector, then our measure of difference
between two networks is the Manhattan distance between the two
vectors. Because there are 24 possible interactions, the maximum
distance between two networks is 24, which occurs if all
interactions that are present on one network are absent in the
other and vice versa.
Identifying good and bad networks and good and bad
combinations of interactions
We were interested in networks that reliably followed a
trajectory from the anterior starting state to the anterior steady
state, as well as from the posterior starting state to the posterior
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
11
September 2010 | Volume 6 | Issue 9 | e1000936
steady state. We defined the overall performance index of a
network, P, as the minimum of the probability of following the
desired anterior trajectory and the probability of following the
desired posterior trajectory. If a network proceeded along each of
the desired trajectories more than 50% of the time, then this was
sufficient to give the average expression gradients observed
experimentally. This is equivalent to Pw0:5. Graphically, we
represent the performance of different networks on plots of
probability of proceeding from the anterior starting state to the
anterior steady state, against probability of proceeding from the
posterior starting state to the posterior steady state (for example,
see Figure 3B). On these plots, networks that reliably produce the
experimentally observed expression gradients (Pw0:5) fall in the
upper, right quadrant.
Finally, we found combinations of interactions that made a
network perform universally poorly or well. We did this by
examining the distribution of P for networks with particular
combinations of interactions. We started by looking at P for all
networks with each single interaction, compared to without. We
then looked at all combinations of interactions being present or
absent for all combinations of two, three and four interactions. If
all the networks containing a particular combination of interac-
tions had Pw0:5, then that set of networks was classified as good.
Conversely, if the majority of networks containing a particular
combination of interactions had Pv0:6, and only a few networks
with 0:5vPv0:6, then that set of networks was classified as bad.
Supporting Information
Table S1
Higher order combinations of interactions that rarely
appear in good networks.
Found at: doi:10.1371/journal.pcbi.1000936.s001 (0.04 MB PDF)
Acknowledgments
We thank Linda Richards and Tomomi Shimogori for their helpful
feedback and comments on the manuscript.
Author Contributions
Conceived and designed the experiments: CEG GJG. Performed the
experiments: CEG. Analyzed the data: CEG. Wrote the paper: CEG GJG.
References
1. Sur M, Leamey CA (2001) Development and plasticity of cortical areas and
networks. Nat Rev Neurosci 2: 251–262.
2. Grove EA, Fukuchi-Shimogori T (2003) Generating the cerebral cortical area
map. Annu Rev Neurosci 26: 355–380.
3. Job C, Tan SS (2003) Constructing the mammalian neocortex: the role of
intrinsic factors. Dev Biol 257: 221–232.
4. Sur M, Rubenstein JLR (2005) Patterning and plasticity of the cerebral cortex.
Science 310: 805–810.
5. O’Leary DDM, Chou SJ, Sahara S (2007) Area patterning of the mammalian
cortex. Neuron 56: 252–269.
6. O’Leary DD, Sahara S (2008) Genetic regulation of arealization of the
neocortex. Curr Opin Neurobiol 18: 90–100.
7. He´bert JM, Fishell G (2008) The genetics of early telencephalon patterning:
some assembly required. Nat Rev Neurosci.
8. Rakic P, Ayoub AE, Breunig JJ, Dominguez MH (2009) Decision by division:
making cortical maps. Trends Neurosci.
9. Shimamura K, Rubenstein JL (1997) Inductive interactions direct early
regionalization of the mouse forebrain. Development 124: 2709–2718.
10. Shimogori T, Banuchi V, Ng HY, Strauss JB, Grove EA (2004) Embryonic
signaling centers expressing BMP, WNT and FGF proteins interact to pattern
the cerebral cortex. Development 131: 5639–5647.
11. Mallamaci A, Stoykova A (2006) Gene networks controlling early cerebral cortex
arealization. Eur J Neurosci 23: 847–856.
12. Wolpert L (1969) Positional information and the spatial pattern of cellular
differentiation. J Theor Biol 25: 1–47.
13. Wolpert L (1996) One hundred years of positional information. Trends Genet
12: 359–364.
14. Zhou C, Qiu Y, Pereira FA, Crair MC, Tsai SY, et al. (1999) The nuclear
orphan receptor COUP-TFI is required for differentiation of subplate neurons
and guidance of thalamocortical axons. Neuron 24: 847–859.
15. Bishop KM, Goudreau G, O’Leary DD (2000) Regulation of area identity in the
mammalian neocortex by Emx2 and Pax6. Science 288: 344–349.
16. Mallamaci A, Muzio L, Chan CH, Parnavelas J, Boncinelli E (2000) Area
identity shifts in the early cerebral cortex of Emx22/2 mutant mice. Nat
Neurosci 3: 679–686.
17. Fukuchi-Shimogori T, Grove EA (2001) Neocortex patterning by the secreted
signaling molecule FGF8. Science 294: 1071–1074.
18. Zhou C, Tsai SY, Tsai MJ (2001) COUP-TFI: an intrinsic factor for early
regionalization of the neocortex. Genes Dev 15: 2054–2059.
19. Bishop KM, Rubenstein JLR, O’Leary DDM (2002) Distinct actions of Emx1,
Emx2, and Pax6 in regulating the specification of areas in the developing
neocortex. J Neurosci 22: 7627–7638.
20. Fukuchi-Shimogori T, Grove EA (2003) Emx2 patterns the neocortex by
regulating FGF positional signaling. Nat Neurosci 6: 825–831.
21. Garel S, Huffman KJ, Rubenstein JLR (2003) Molecular regionalization of the
neocortex is disrupted in Fgf8 hypomorphic mutants. Development 130:
1903–1914.
22. Hamasaki T, Leinga¨rtner A, Ringstedt T, O’Leary DDM (2004) EMX2
regulates sizes and positioning of the primary sensory and motor areas in
neocortex by direct specification of cortical progenitors. Neuron 43: 359–372.
23. Armentano M, Chou SJ, Tomassy GS, Leinga¨rtner A, O’Leary DDM, et al.
(2007) COUP-TFI regulates the balance of cortical patterning between frontal/
motor and sensory areas. Nat Neurosci 10: 1277–1286.
24. Sahara S, Kawakami Y, Belmonte JCI, O’Leary DDM (2007) Sp8 exhibits
reciprocal induction with Fgf8 but has an opposing effect on anterior-posterior
cortical area patterning. Neural Develop 2: 10.
25. Leinga¨rtner A, Thuret S, Kroll TT, Chou SJ, Leasure JL, et al. (2007) Cortical
area size dictates performance at modality-specific behaviors. Proc Natl Acad
Sci U S A 104: 4153–4158.
26. Zembrzycki A, Griesel G, Stoykova A, Mansouri A (2007) Genetic interplay
between the transcription factors Sp8 and Emx2 in the patterning of the
forebrain. Neural Develop 2: 8.
27. Cholfin JA, Rubenstein JLR (2008) Frontal cortex subdivision patterning is
coordinately regulated by Fgf8, Fgf17, and Emx2. J Comp Neurol 509: 144–155.
28. Faedo A, Tomassy GS, Ruan Y, Teichmann H, Krauss S, et al. (2008) COUP-
TFI coordinates cortical patterning, neurogenesis, and laminar fate and
modulates MAPK/ERK, AKT, and beta-catenin signaling. Cereb Cortex 18:
2117–2131.
29. Pin˜on MC, Tuoc TC, Ashery-Padan R, Molna´r Z, Stoykova A (2008) Altered
molecular regionalization and normal thalamocortical connections in cortex-
specific Pax6 knock-out mice. J Neurosci 28: 8724–8734.
30. Manuel M, Georgala PA, Carr CB, Chanas S, Kleinjan DA, et al. (2007)
Controlled overexpression of Pax6 in vivo negatively autoregulates the Pax6
locus, causing cell-autonomous defects of late cortical progenitor proliferation
with little effect on cortical arealization. Development 134: 545–555.
31. Davidson EH, Erwin DH (2006) Gene regulatory networks and the evolution of
animal body plans. Science 311: 796–800.
32. Bolouri H (2008) Embryonic pattern formation without morphogens. Bioessays
30: 412–417.
33. Smolen P, Baxter DA, Byrne JH (2000) Mathematical modeling of gene
networks. Neuron 26: 567–580.
34. Smolen P, Baxter DA, Byrne JH (2000) Modeling transcriptional control in gene
networks–methods, recent results, and future directions. Bull Math Biol 62:
247–292.
35. de Jong H (2002) Modeling and simulation of genetic regulatory systems: a
literature review. J Comput Biol 9: 67–103.
36. Klipp E, Liebermeister W (2006) Mathematical modeling of intracellular
signaling pathways. BMC Neurosci 7 Suppl 1: S10.
37. Lewis J (2008) From signals to patterns: space, time, and mathematics in
developmental biology. Science 322: 399–403.
38. Lazebnik Y (2003) Can a biologist fix a radio, or what I learned while studying
apoptosis. Cancer Cell 2: 179–182.
39. Hasty J, McMillen D, Collins JJ (2002) Engineered gene circuits. Nature 420:
224–230.
40. Thieffry D, Sanchez L (2003) Dynamical modelling of pattern formation during
embryonic development. Curr Opin Genet Dev 13: 326–330.
41. Tomlin CJ, Axelrod JD (2007) Biology by numbers: mathematical modelling in
developmental biology. Nat Rev Genet 8: 331–340.
42. Fisher J, Henzinger TA (2007) Executable cell biology. Nat Biotechnol 25:
1239–1249.
43. Karlebach G, Shamir R (2008) Modelling and analysis of gene regulatory
networks. Nat Rev Mol Cell Biol 9: 770–780.
44. Kestler HA, Wawra C, Kracher B, Ku¨hl M (2008) Network modeling of signal
transduction: establishing the global view. Bioessays 30: 1110–1125.
45. Bolouri H (2008) Computational Modeling of Gene Regulatory Networks — a
Primer. London: Imperial College Press.
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
12
September 2010 | Volume 6 | Issue 9 | e1000936
46. Bhalla US, Iyengar R (1999) Emergent properties of networks of biological
signaling pathways. Science 283: 381–387.
47. Bornholdt S (2008) Boolean network models of cellular regulation: prospects and
limitations. J R Soc Interface 5 Suppl 1: S85–S94.
48. Nakajima A, Isshiki T, Kaneko K, Ishihara S (2010) Robustness under
functional constraint: the genetic network for temporal expression in drosophila
neurogenesis. PLoS Comput Biol 6: e1000760.
49. Thomas R (1991) Regulatory networks seen as asynchronous automata: a logical
description. J Theor Biol 153: 1–23.
50. Bolouri H, Davidson EH (2002) Modeling transcriptional regulatory networks.
Bioessays 24: 1118–1129.
51. Davidson EH, Levine MS (2008) Properties of developmental gene regulatory
networks. Proc Natl Acad Sci U S A 105: 20063–20066.
52. Crossley PH, Martinez S, Ohkubo Y, Rubenstein JL (2001) Coordinate
expression of Fgf8, Otx2, Bmp4, and Shh in the rostral prosencephalon during
development of the telencephalic and optic vesicles. Neuroscience 108: 183–206.
53. Storm EE, Rubenstein JLR, Martin GR (2003) Dosage of Fgf8 determines
whether cell survival is positively or negatively regulated in the developing
forebrain. Proc Natl Acad Sci U S A 100: 1757–1762.
54. Storm EE, Garel S, Borello U, Hebert JM, Martinez S, et al. (2006) Dose-
dependent functions of Fgf8 in regulating telencephalic patterning centers.
Development 133: 1831–1844.
55. Muzio L, DiBenedetto B, Stoykova A, Boncinelli E, Gruss P, et al. (2002) Emx2
and Pax6 control regionalization of the pre-neuronogenic cortical primordium.
Cereb Cortex 12: 129–139.
56. Muzio L, Mallamaci A (2003) Emx1, Emx2 and Pax6 in specification,
regionalization and arealization of the cerebral cortex. Cereb Cortex 13:
641–647.
57. Crossley PH, Martinez S, Martin GR (1996) Midbrain development induced by
FGF8 in the chick embryo. Nature 380: 66–68.
58. Crossley PH, Minowada G, MacArthur CA, Martin GR (1996) Roles for FGF8
in the induction, initiation, and maintenance of chick limb development. Cell 84:
127–136.
59. Karbowski J, Ermentrout GB (2004) Model of the early development of
thalamo-cortical connections and area patterning via signaling molecules.
J Comput Neurosci 17: 347–363.
60. Schlitt T, Brazma A (2007) Current approaches to gene regulatory network
modelling. BMC Bioinformatics 8 Suppl 6: S9.
61. Laubenbacher R, Stigler B (2004) A computational algebra approach to the
reverse engineering of gene regulatory networks. J Theor Biol 229: 523–537.
62. Wittmann DM, Blchl F, Trmbach D, Wurst W, Prakash N, et al. (2009) Spatial
analysis of expression patterns predicts genetic interactions at the mid-hindbrain
boundary. PLoS Comput Biol 5: e1000569.
63. Albert R, Othmer HG (2003) The topology of the regulatory interactions
predicts the expression pattern of the segment polarity genes in Drosophila
melanogaster. J Theor Biol 223: 1–18.
64. Hallinan J, Wiles J (2004) Asynchronous dynamics of an artificial genetic
regulatory network. In: Pollack J, Bedau M, Husbands P, Ikegami T,
Watson RA, eds. Artificial life IX: Proceedings of the ninth international
conference on the simulation and synthesis of living systems. CambridgeMA:
MIT Press. pp 399–403.
65. Gebali F Reducible Markov Chains, Springer, chapter 5. pp 151–182.
doi:10.1007/978-0-387-74437-7.
66. Kauffman SA (1993) The Origins of Order. New York: Oxford University Press.
Gene Regulation of Brain Development
PLoS Computational Biology | www.ploscompbiol.org
13
September 2010 | Volume 6 | Issue 9 | e1000936
|
20862356
|
Sp8 = ( ( Fgf8 ) AND NOT ( Emx2 ) )
Coup_fti = ( NOT ( ( Fgf8 ) OR ( Sp8 ) ) ) OR NOT ( Fgf8 OR Sp8 )
Pax6 = ( ( ( Sp8 ) AND NOT ( Coup_fti ) ) AND NOT ( Emx2 ) )
Fgf8 = ( ( Fgf8 AND ( ( ( Sp8 ) ) ) ) AND NOT ( Emx2 ) )
Emx2 = ( ( ( ( Coup_fti ) AND NOT ( Fgf8 ) ) AND NOT ( Sp8 ) ) AND NOT ( Pax6 ) )
|
Dynamical and Structural Analysis of a T Cell Survival
Network Identifies Novel Candidate Therapeutic Targets
for Large Granular Lymphocyte Leukemia
Assieh Saadatpour1, Rui-Sheng Wang2, Aijun Liao3, Xin Liu3, Thomas P. Loughran3, Istva´n Albert4, Re´ka
Albert2*
1 Department of Mathematics, The Pennsylvania State University, University Park, Pennsylvania, United States of America, 2 Department of Physics, The Pennsylvania State
University, University Park, Pennsylvania, United States of America, 3 Penn State Hershey Cancer Institute, The Pennsylvania State University College of Medicine, Hershey,
Pennsylvania, United States of America, 4 Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United
States of America
Abstract
The blood cancer T cell large granular lymphocyte (T-LGL) leukemia is a chronic disease characterized by a clonal
proliferation of cytotoxic T cells. As no curative therapy is yet known for this disease, identification of potential therapeutic
targets is of immense importance. In this paper, we perform a comprehensive dynamical and structural analysis of a
network model of this disease. By employing a network reduction technique, we identify the stationary states (fixed points)
of the system, representing normal and diseased (T-LGL) behavior, and analyze their precursor states (basins of attraction)
using an asynchronous Boolean dynamic framework. This analysis identifies the T-LGL states of 54 components of the
network, out of which 36 (67%) are corroborated by previous experimental evidence and the rest are novel predictions. We
further test and validate one of these newly identified states experimentally. Specifically, we verify the prediction that the
node SMAD is over-active in leukemic T-LGL by demonstrating the predominant phosphorylation of the SMAD family
members Smad2 and Smad3. Our systematic perturbation analysis using dynamical and structural methods leads to the
identification of 19 potential therapeutic targets, 68% of which are corroborated by experimental evidence. The novel
therapeutic targets provide valuable guidance for wet-bench experiments. In addition, we successfully identify two new
candidates for engineering long-lived T cells necessary for the delivery of virus and cancer vaccines. Overall, this study
provides a bird’s-eye-view of the avenues available for identification of therapeutic targets for similar diseases through
perturbation of the underlying signal transduction network.
Citation: Saadatpour A, Wang R-S, Liao A, Liu X, Loughran TP, et al. (2011) Dynamical and Structural Analysis of a T Cell Survival Network Identifies Novel
Candidate Therapeutic Targets for Large Granular Lymphocyte Leukemia. PLoS Comput Biol 7(11): e1002267. doi:10.1371/journal.pcbi.1002267
Editor: Yanay Ofran, Bar Ilan University, Israel
Received May 18, 2011; Accepted September 22, 2011; Published November 10, 2011
Copyright: 2011 Saadatpour et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work was supported by NSF grant CCF-0643529 to RA and by NIH grants CA98472 and CA133525 to TPL. The funders had no role in study design,
data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: ralbert@phys.psu.edu
Introduction
Living cells perceive and respond to environmental perturba-
tions in order to maintain their functional capabilities, such as
growth, survival, and apoptosis. This process is carried out
through a cascade of interactions forming complex signaling
networks. Dysregulation (abnormal expression or activity) of some
components in these signaling networks affects the efficacy of
signal transduction and may eventually trigger a transition from
the normal physiological state to a dysfunctional system [1]
manifested as diseases such as diabetes [2,3], developmental
disorders [4], autoimmunity [5] and cancer [4,6]. For example,
the blood cancer T-cell large granular lymphocyte (T-LGL)
leukemia exhibits an abnormal proliferation of mature cytotoxic T
lymphocytes (CTLs). Normal CTLs are generated to eliminate
cells infected by a virus, but unlike normal CTLs which undergo
activation-induced cell death after they successfully fight the virus,
leukemic T-LGL cells remain long-term competent [7]. The cause
of this abnormal behavior has been identified as dysregulation of a
few components of the signal transduction network responsible for
activation-induced cell death in T cells [8].
Network representation, wherein the system’s components are
denoted as nodes and their interactions as edges, provides a
powerful tool for analyzing many complex systems [9,10,11]. In
particular, network modeling has recently found ever-increasing
applications in understanding the dynamic behavior of intracel-
lular biological systems in response to environmental stimuli and
internal perturbations [12,13,14]. The paucity of knowledge on
the biochemical kinetic parameters required for continuous
models has called for alternative dynamic approaches. Among
the most successful approaches are discrete dynamic models in
which each component is assumed to have a finite number of
qualitative states, and the regulatory interactions are described by
logical functions [15]. The simplest discrete dynamic models are
the so-called Boolean models that assume only two states (ON or
OFF)
for
each
component.
These
models
were
originally
introduced by S. Kauffman and R. Thomas to provide a coarse-
grained description of gene regulatory networks [16,17].
PLoS Computational Biology | www.ploscompbiol.org
1
November 2011 | Volume 7 | Issue 11 | e1002267
A Boolean network model of T cell survival signaling in the
context of T-LGL leukemia was previously constructed by Zhang
et al [18] through performing an extensive literature search. This
network consists of 60 components, including proteins, mRNAs,
and small molecules (see Figure 1). The main input to the network
is ‘‘Stimuli’’, which represents virus or antigen stimulation, and the
main output node is ‘‘Apoptosis’’, which denotes programmed cell
death. Based on a random order asynchronous Boolean dynamic
model of the assembled network, Zhang et al identified a minimal
number of dysregulations that can cause the T-LGL survival state,
namely overabundance or overactivity of the proteins platelet-
derived growth factor (PDGF) and interleukin 15 (IL15). Zhang
et al carried out a preliminary analysis of the network’s dynamics
by performing numerical simulations starting from one specific
initial condition (corresponding to resting T cells receiving antigen
stimulation and over-abundance of the two proteins PDGF and
IL15). Once the known deregulations in T-LGL leukemia were
reproduced, each of these deregulations was interrupted individ-
ually, by setting the node’s status to the opposite state, to predict
key mediators of the disease. Yet, a complete dynamic analysis of
the system, including identification of the attractors (e.g. steady
states) of the system and their corresponding basin of attraction
(precursor states), as well as a thorough perturbation analysis of the
system considering all possible initial states, is lacking. Performing
this analysis can provide deeper insights into unknown aspects of
T-LGL leukemia.
Stuck-at-ON/OFF fault is a very common dysregulation of
biomolecules in various cancer diseases [19]. For example, stuck-
at-ON (constitutive activation) of the RAS protein in the mitogen-
activated protein kinase pathways leads to aberrant cell prolifer-
ation and cancer [19,20]. Thus identifying components whose
stuck-at values result in the clearance, or alternatively, the
persistence of a disease is extremely beneficial for the design of
intervention strategies. As there is no known curative therapy for
T-LGL leukemia, identification of potential therapeutic targets is
of utmost importance [21].
In this paper, we carry out a detailed analysis of the T-LGL
signaling network by considering all possible initial states to probe
the long-term behavior of the underlying disease. We employ an
asynchronous
Boolean
dynamic
framework
and
a
network
reduction method, which we previously proposed [22], to identify
the attractors of the system and analyze their basins of attraction.
This analysis allows us to confirm or predict the T-LGL states of
54 components of the network. The predicted state of one of the
components (SMAD) is validated by new wet-bench experiments.
We then perform node perturbation analysis using the dynamic
approach and a structural method proposed in [23] to study to
what extent does each component contribute to T-LGL leukemia.
Both methods give consistent results and together identify 19 key
components whose disruption can reverse the abnormal state of
the signaling network, thereby uncovering potential therapeutic
targets for this disease, some of which are also corroborated by
experimental evidence.
Materials and Methods
Any biological regulatory network can be represented by a
directed graph G = (V, E) where V = {v1, v2,…, vn} is the set of
vertices (nodes) describing different components of the system, and
E is the set of edges denoting the regulatory interactions among the
components. The orientation of each edge in the network follows
the direction of mass transfer or information propagation from the
upstream to the downstream node. Each edge can be also
characterized with a sign where a positive sign denotes activation
and a negative sign signifies inhibition. The source nodes (i.e.
nodes with no incoming edges) of this graph, if they exist, represent
external inputs (signals), and one or more nodes, usually sink nodes
(i.e. nodes with no outgoing edges), are customarily designated as
outputs of the network.
Boolean dynamic models
Boolean models belong to the class of discrete dynamic models
in which each node of the network is characterized by an ON (1)
or OFF (0) state and usually the time variable t is also considered to
be discrete, i.e. it takes nonnegative integer values [24,25]. The
future state of each node vi is determined by the current states of
the nodes regulating it according to a Boolean transfer function
fi : f0,1gki?f0,1g, where ki is the number of regulators of vi. Each
Boolean function (rule) represents the regulatory relationships
between the components and is usually expressed via the logical
operators AND, OR and NOT. The state of the system at each
time step is denoted by a vector whose ith component represents
the state of node vi at that time step. The discrete state space of a
system can be represented by a state transition graph whose nodes
are states of the system and edges are allowed transitions among
the states. By updating the nodes’ states at each time step, the state
of the system evolves over time and following a trajectory of states
it eventually settles down into an attractor. An attractor can be in
the form of either a fixed point, in which the state of the system
does not change, or a complex attractor, where the system
oscillates (regularly or irregularly) among a set of states. The set of
states leading to a specific attractor is called the basin of attraction
of that attractor.
In order to evaluate the state of each node at a given time
instant, synchronous as well as asynchronous updating strategies
have been proposed [24,25]. In the synchronous method all nodes
of the network are updated simultaneously at multiples of a
common time step. The underlying assumption of this update
method is that the timescales of all the processes occurring in a
system are similar. This is a quite strong and potentially unrealistic
assumption, which in particular may not be suited for intracellular
biological processes due to the variety of timescales associated with
transcription, translation and post-translational mechanisms [26].
To overcome this limitation, various asynchronous methods have
been proposed wherein the nodes are updated based on individual
Author Summary
T-LGL leukemia is a blood cancer characterized by an
abnormal increase in the abundance of a type of white
blood cell called T cell. Since there is no known curative
therapy for this disease, identification of potential thera-
peutic targets is of utmost importance. Experimental
identification of manipulations capable of reversing the
disease condition is usually a long, arduous process.
Mathematical modeling can aid this process by identifying
potential therapeutic interventions. In this work, we carry
out a systematic analysis of a network model of T cell
survival in T-LGL leukemia to get a deeper insight into the
unknown facets of the disease. We identify the T-LGL
status of 54 components of the system, out of which 36
(67%) are corroborated by previous experimental evidence
and the rest are novel predictions, one of which we
validate by follow-up experiments. By deciphering the
structure and dynamics of the underlying network, we
identify
component
perturbations that
lead to pro-
grammed cell death, thereby suggesting several novel
candidate therapeutic targets for future experiments.
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
2
November 2011 | Volume 7 | Issue 11 | e1002267
timescales [25,27,28,29,30], including deterministic methods with
fixed node timescales and stochastic methods such as random
order asynchronous method [27] wherein the nodes are updated
in random permutations. In a previous work [22], we carried out a
comparative study of three different asynchronous methods
applied to the same biological system. That study suggested that
the general asynchronous (GA) method, wherein a randomly
selected node is updated at each time step, is the most efficient and
informative asynchronous updating strategy. This is because
deterministic asynchronous [22] or autonomous [30] Boolean
models require kinetic or timing knowledge, which is usually
missing, and random order asynchronous models [27] are not
computationally efficient compared to the GA models. In addition,
the superiority of the GA approach has been corroborated by
other researchers [29] and the method has been used in other
studies as well [31,32]. We thus chose to employ the GA method
in this work, and we implemented it using the open-source
software library BooleanNet [33]. It is important to note that the
stochasticity inherent to this method may cause each state to have
multiple successors, and thus the basins of attraction of different
attractors may overlap. For systems with multiple fixed-point
attractors, the absorption probabilities to each fixed point can be
computed through the analysis of the Markov chain and transition
matrix associated with the state transition graph of the system [34].
Given a fixed point, node perturbations can be performed by
reversing the state of the nodes i.e. by knocking out the nodes that
stabilize in an ON state in the fixed point or over-expressing the
ones that stabilize in an OFF state.
Figure 1. The T-LGL survival signaling network. The shape of the nodes indicates the cellular location: rectangular indicates intracellular
components, ellipse indicates extracellular components, and diamond indicates receptors. Node colors reflect the current knowledge on the state of
these nodes in leukemic cells: highly active components in T-LGL are shown in red, inhibited nodes are shown in green, nodes that have been
suggested to be deregulated are in blue, and the state of white nodes is unknown. Conceptual nodes (Stimuli, Stimuli2, P2, Cytoskeleton signaling,
Proliferation, and Apoptosis) are represented by yellow hexagons. An arrowhead or a short perpendicular bar at the end of an edge indicates
activation or inhibition, respectively. The inhibitory edges from Apoptosis to other nodes are not shown. The full names of the node labels are given
in Table S2. This figure and its caption are adapted from [18].
doi:10.1371/journal.pcbi.1002267.g001
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
3
November 2011 | Volume 7 | Issue 11 | e1002267
Network reduction
A Boolean network with n nodes has a total of 2n states. This
exponential dependence makes it computationally intractable to
map the state transition graphs of even relatively small networks.
This calls for developing efficient network reduction approaches.
Recent efforts towards addressing this challenge consists of iteratively
removing single nodes that do not regulate their own function and
simplifying the redundant transfer functions using Boolean algebra
[35,36]. Naldi et al [35] proved that this approach preserves the fixed
points of the system and that for each (irregular) complex attractor in
the original asynchronous model there is at least one complex
attractor in the reduced model (i.e. network reduction may create
spurious oscillations). Boolean networks often contain nodes whose
states stabilize in an attracting state after a transient period,
regardless of updating strategy or initial conditions. The attracting
states of these nodes can be readily identified by inspection of their
Boolean functions. In a previous work [22] we proposed a method of
network simplification by (i) pinpointing and eliminating these
stabilized nodes and (ii) iteratively removing a simple mediator node
(e.g. a node that has one incoming edge and one outgoing edge) and
connecting its input(s) to its target(s). Our simplification method
shares similarities with the method proposed in [35,36], with the
difference that we only remove stabilized nodes (which have the
same state on every attractor) and simple mediator nodes rather than
eliminating each node without a self loop. Thus their proof regarding
the preservation of the steady states by the reduction method holds
true in our case. We employed this simplification method for the
analysis of a signal transduction network in plants and verified by
using numerical simulations that it preserves the attractors of that
system. In this work, we employ this reduction method to simplify the
T-LGL leukemia signal transduction network synthesized by Zhang
et al [18], thereby facilitating its dynamical analysis. We also note that
the first step of our simplification method is similar to the logical
steady state analysis implemented in the software tool CellNetAna-
lyzer [37,38]. We thus refer to this step as logical steady state analysis
throughout the paper.
Identification of attractors
It should be noted that the fixed points of a Boolean network are
the same for both synchronous and asynchronous methods. In
order to obtain the fixed points of a system one can solve the set of
Boolean equations independent of time. To this end, we first fix
the state of the source nodes. We then determine the nodes whose
rules depend on the source nodes and will either stabilize in an
attracting state after a time delay or otherwise their rules can be
simplified significantly by plugging in the state of the source nodes.
Iteratively inserting the states of stabilized nodes in the rules (i.e.
employing logical steady state analysis) will result in either the
fixed point(s) of the system, or the partial fixed point(s) and a
remaining set of equations to be solved. In the latter case, if the
remaining set of equations is too large to obtain its fixed point(s)
analytically, we take advantage of the second step of our reduction
method [22] to simplify the resulting network and to determine a
simpler set of Boolean rules. By solving this simpler set of
equations (or performing numerical simulations, if necessary) and
plugging the solutions into the original rules, we can then find the
states of the removed nodes and determine the attractors of the
whole system accordingly. For the analysis of basins of attraction
of the attractors, we perform numerical simulations using the GA
update method.
A structural method for identifying essential components
The topology (structure) and the function of biological networks
are closely related. Therefore, structural analysis of biological
networks provides an alternative way to understand their function
[39,40]. We have recently proposed an integrative method to
identify the essential components of any given signal transduction
network [23]. The starting point of the method is to represent the
combinatorial relationship of multiple regulatory interactions
converging on a node v by a Boolean rule:
v~(u11 AND ::: AND u1n1) OR (u21 AND ::: AND u2n2)
OR ::: OR (um1 AND ::: AND umnm)
where uij’s are regulators of node v. The method consists of two
main steps. The first step is the expansion of a signaling network to
a new representation by incorporating the sign of the interactions
as well as the combinatorial nature of multiple converging
interactions. This is achieved by introducing a complementary
node for each component that plays a role in negative regulations
(NOT operation) as well as introducing a composite node to
denote conditionality among two or more edges (AND operation).
This step eliminates the distinction of the edge signs; that is, all
directed edges in the expanded network denote activation. In
addition, the AND and OR operators can be readily distinguished
in the expanded network, i.e., multiple edges ending at composite
nodes are added by the AND operator, while multiple edges
ending at original or complementary nodes are cumulated by the
OR operator. The second step is to model the cascading effects
following the loss of a node by an iterative process that identifies
and removes nodes that have lost their indispensable regulators.
These two steps allow ranking of the nodes by the effects of their
loss on the connectivity between the network’s input(s) and
output(s). We proposed two connectivity measures in [23], namely
the simple path (SP) measure, which counts the number of all
simple paths from inputs to outputs, and a graph measure based
on elementary signaling modes (ESMs), defined as a minimal set of
components that can perform signal transduction from initial
signals to cellular responses. We found that the combinatorial
aspects of ESMs pose a substantial obstacle to counting them in
large networks and that the SP measure has a similar performance
as the ESM measure since both measures incorporate the
cascading effects of a node’s removal arising from the synergistic
relations between multiple interactions. Therefore, we employ the
SP measure and define the importance value of a component v as:
ESP(v)~ NSP(Gexp){NSP(GDv)
NSP(Gexp)
where NSP(Gexp) and NSP(GDv) denote the total number of simple
paths from the input(s) to the output(s) in the original expanded
network Gexp and the damaged network GDv upon disruption of node
v, respectively. This essentiality measure takes values in the interval
[0,1], with 1 indicating a node whose loss causes the disruption of all
paths between the input and output node(s). In this paper, we also
make use of this structural method to identify essential components
of the T-LGL leukemia signaling network. We then relate the
importance value of nodes to the effects of their knockout (sustained
OFF state) in the dynamic model and the importance value of
complementary nodes to the effects of their original nodes’
constitutive activation (sustained ON state) in the dynamic model.
Experimental determination of the T-LGL state of the
node SMAD
Patient characteristics and preparation of peripheral
blood mononuclear cells (PBMC).
All patients met the
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
4
November 2011 | Volume 7 | Issue 11 | e1002267
clinical criteria of T-LGL leukemia with increased numbers
(.80%) of CD3+CD8+ T cells in the peripheral blood. Patients
received no treatment at the time of sample acquisition. Peripheral
blood specimens from LGL leukemia patients were obtained and
informed consents signed for sample collection according to a
protocol approved by the Institutional Review Board of Penn State
Hershey Cancer Institute. PBMC were isolated by Ficoll-Hypaque
gradient separation, as described previously [41]. CD3+CD8+ T
cells from four age- and gender-matched healthy donors were
isolated by a human CD8+ T cell enrichment cocktail RosetteSep
kit
(Stemcell
Technology).
The
purity
of
freshly
isolated
CD3+CD8+ T cells (26105/sample in triplicate) in each of the
samples was determined by flow cytometry assay by detecting
positive staining of the CD3 and CD8 T cell markers. The purity
for normal purified CD3+CD8+ T cells was over 90%. Cell
viability was determined by trypan blue exclusion assay with more
than 95% viability in all the samples.
Phospho-Smad2
and
phospho-Smad3
measurement.
Western blot was performed to detect Phospho-Smad2 (P-
Smad2) and Phospho-Smad3 (P-Smad3) in activated normal
CD3+CD8+ cells (CD3+CD8+ cells .90%) compared with PBMC
(CD3+CD8+ cells .80%) from T-LGL leukemia patients. Normal
CD3+CD8+ T cells were isolated by a human CD8+ T cell
enrichment cocktail RosetteSep kit (Stemcell Technology) from
four normal donors, then cultured in RPMI-1640 supplemented
with 10% fetal bovine serum in presence of PHA (1 mg/mL) for 1
day followed by IL2 (500 IU/mL) for 3 days (lanes 1–4). The
equal loading of protein was confirmed by probing with total
Smad2
or
Smad3.
Phospho-Smad2
(Ser465/467),
Smad2,
Phospho-Smad3
(Ser423/425)
and
Smad3
antibodies
were
purchased from Cell Signaling Technology Inc. (Beverly, MA).
Results
Network simplification and dynamic analysis
The T-LGL signaling network reconstructed by Zhang et al [18]
contains 60 nodes and 142 regulatory edges. Zhang et al used a
two-step process: they first synthesized a network containing 128
nodes and 287 edges by extensive literature search, then simplified
it with the software NET-SYNTHESIS [42], which constructs the
sparsest network that maintains all of the causal (upstream-
downstream) effects incorporated in a redundant starting network.
In this study, we work with the 60-node T-LGL signaling network
reported in [18], which is redrawn in Figure 1. The Boolean rules
for the components of the network were constructed in [18] by
synthesizing experimental observations and for convenience are
given in Table S1 as well. The description of the node names and
abbreviations are provided in Table S2.
To reduce the computational burden associated with the large
state space (more than 1018 states for 60 nodes), we simplified the
T-LGL network using the reduction method proposed in [22] (see
Materials and Methods). We fixed the six source nodes in the states
given in [18], i.e. Stimuli, IL15, and PDGF were fixed at ON and
Stimuli2, CD45, and TAX were fixed at OFF. We used the
Boolean rules constructed in [18], with one notable difference.
The Boolean rules for all the nodes in [18], except Apoptosis,
contain the expression ‘‘AND NOT Apoptosis’’, meaning that if
Apoptosis is ON, the cell dies and correspondingly all other nodes
are turned OFF. To focus on the trajectory leading to the initial
turning on of the Apoptosis node, we removed the ‘‘AND NOT
Apoptosis’’ from all the logical rules. This allows us to determine
the stationary states of the nodes in a live cell. We determined
which
nodes’
states
stabilize
using
the
first
step
of
our
simplification method, i.e. logical steady state analysis (see
Materials and Methods). Our analysis revealed that 36 nodes of
the network stabilize in either an ON or OFF state. In particular,
Proliferation and Cytoskeleton signaling, two output nodes of the
network, stabilize in the OFF and ON state, respectively. Low
proliferation in leukemic LGL has been observed experimentally
[43], which supports our finding of a long-term OFF state for this
output node. The ON state of Cytoskeleton signaling may not be
biologically relevant as this node represents the ability of T cells to
attach and move which is expected to be reduced in leukemic T-
LGL compared to normal T cells. The nodes whose stabilized
states cannot be readily obtained by inspection of their Boolean
rules form the sub-network represented in Figure 2A. The Boolean
rules of these nodes are listed in Table S3 wherein we put back the
‘‘AND NOT Apoptosis’’ expression into the rules.
Next, we identified the attractors (long-term behavior) of the
sub-network
represented
in
Figure
2A
(see
Materials
and
Methods). We found that upon activation of Apoptosis all other
nodes stabilize at OFF, forming the normal fixed point of the
system, which represents the normal behavior of programmed cell
death. When Apoptosis is stabilized at OFF, the two nodes in the
top sub-graph oscillate while all the nodes in the bottom sub-graph
are stabilized at either ON or OFF. As shown in Figure 3, the state
space of the two oscillatory nodes, TCR and CTLA4, forms a
complex attractor in which the average fraction of ON states for
either node is 0.5. Given that these two nodes have no effect on
any other node under the conditions studied here (i.e. stable states
of the source nodes), their behavior can be separated from the rest
of the network.
The bottom sub-graph exhibits the normal fixed point, as well
as two T-LGL (disease) fixed points in which Apoptosis is OFF.
The only difference between the two T-LGL fixed points is that
the node P2 is ON in one fixed point and OFF in the other, which
was expected due to the presence of a self-loop on P2 in Figure 2A.
P2 is a virtual node introduced to mediate the inhibition of
interferon-c translation in the case of sustained activity of the
interferon-c protein (IFNG in Figure 2A). The node IFNG is also
inhibited by the node SMAD which stabilizes in the ON state in
both T-LGL fixed points. Therefore IFNG stabilizes at OFF,
irrespective of the state of P2, as supported by experimental
evidence [44]. Thus the biological difference between the two
fixed points is essentially a memory effect, i.e. the ON state of P2
indicates that IFNG was transiently ON before stabilizing in the
OFF state. In the two T-LGL fixed points for the bottom sub-
graph of Figure 2A, the nodes sFas, GPCR, S1P, SMAD, MCL1,
FLIP, and IAP are ON and the other nodes are OFF. We found
by numerical simulations using the GA method (see Materials and
Methods) that out of 65,536 total states in the state transition
graph, 53% are in the exclusive basin of attraction of the normal
fixed point, 0.24% are in the exclusive basin of attraction of the T-
LGL fixed point wherein P2 is ON and 0.03% are in the exclusive
basin of attraction of the T-LGL fixed point wherein P2 is OFF.
Interestingly, there is a significant overlap among the basins of
attraction of all the three fixed points. The large basin of attraction
of the normal fixed point is partly due to the fact that all the states
having Apoptosis in the ON state (that is, half of the total number
of states) belong to the exclusive basin of the normal fixed point.
These states are not biologically relevant initial conditions but they
represent potential intermediary states toward programmed cell
death and as such they need to be included in the state transition
graph.
Since the state transition graph of the bottom sub-graph given
in Figure 2A is too large to represent and to further analyze (e.g. to
obtain the probabilities of reaching each of the fixed points), we
applied the second step of the network reduction method proposed
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
5
November 2011 | Volume 7 | Issue 11 | e1002267
in [22]. This step preserves the fixed points of the system (see
Materials and Methods), and since the only attractors of this sub-
graph are fixed points, the state space of the reduced network is
expected to reflect the properties of the full state space.
Correspondingly, the nodes having in-degree and out-degree of
one (or less) in the sub-graph on Figure 2A, such as sFas, MCL1,
IAP, GPCR, SMAD, and CREB, can be safely removed without
losing any significant information as such nodes at most introduce
a delay in the signal propagation. In addition, we note that
although the node P2 has a self-loop and generates a new T-LGL
fixed point as described before, it can also be removed from the
network since the two fixed points differ only in the state of P2 and
thus correspond to biologically equivalent disease states. We revisit
this node when enumerating the attractors of the original network.
In the resulting simplified network, the nodes BID, Caspase, and
IFNG would also have in-degree and out-degree of one (or less)
and thus can be safely removed as well. This reduction procedure
results in a simple sub-network represented in Figure 2B with the
Boolean rules given in Table 1.
Our attractor analysis revealed that this sub-network has two
fixed points, namely 000001 and 110000 (the digits from left to
right represent the state of the nodes in the order as listed from top
to bottom in Table 1). The first fixed point represents the normal
state, that is, the apoptosis of CTL cells. Note that the OFF state of
other nodes in this fixed point was expected because of the
presence of ‘‘AND NOT Apoptosis’’ in all the Boolean rules. The
second fixed point is the T-LGL (disease) one as Apoptosis is
stabilized in the OFF state. We note that the sub-network depicted
in Figure 2B contains a backbone of activations from Fas to
Apoptosis and two nodes (S1P and FLIP) which both have a
mutual inhibitory relationship with the backbone. If activation
reaches Apoptosis, the system converges to the normal fixed point.
In the T-LGL fixed point, on the other hand, the backbone is
inactive while S1P and FLIP are active.
We found by simulations that for the simplified network of
Figure 2B, 56% of the states of the state transition graph
(represented in Figure 4) are in the exclusive basin of attraction of
the normal fixed point while 5% of the states form the exclusive
basin of attraction of the T-LGL fixed point. Again, the half of
state space that has the ON state of Apoptosis belongs to the
exclusive basin of attraction of the normal fixed point. Notably,
there is a significant overlap between the basins of attraction of the
two fixed points, which is illustrated by a gray color in Figure 4.
The probabilities of reaching each of the two fixed points starting
from
these
gray-colored
states,
found
by
analysis
of
the
corresponding Markov chain (see Materials and Methods), are
given in Figure 5. As this figure represents, for the majority of cases
the probability of reaching the normal fixed point is higher than
that of the T-LGL fixed point. The three states whose probabilities
to reach the T-LGL fixed point are greater than or equal to 0.7 are
one step away either from the T-LGL fixed point or from the
states in its exclusive basin of attraction. In two of them, the
Figure 2. Reduced sub-networks of the T-LGL signaling network. The full names of the nodes can be found in Table S2. An arrowhead or a
short perpendicular bar at the end of an edge indicates activation or inhibition, respectively. The inhibitory edges from Apoptosis to other nodes are
not shown. (A) The 18-node sub-network. This sub-network is obtained by removing the nodes that stabilize in the ON or OFF state upon fixing
the state of the source nodes. (B) The 6-node sub-network. This sub-network is obtained by removing the top sub-graph of the sub-network in
(A) and merging simple mediator nodes in the bottom sub-graph.
doi:10.1371/journal.pcbi.1002267.g002
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
6
November 2011 | Volume 7 | Issue 11 | e1002267
backbone of the network in Figure 2B is inactive, and in the third
one the backbone is partially inactive and most likely will remain
inactive due to the ON state of S1P (one of the two nodes having
mutual inhibition with the backbone).
Based on the sub-network analysis and considering the states of
the nodes that stabilized at the beginning based on the logical
steady state analysis, we conclude that the whole T-LGL network
has three attractors, namely the normal fixed point wherein
Apoptosis is ON and all other nodes are OFF, representing the
normal physiological state, and two T-LGL attractors in which all
nodes except two, i.e. TCR and CTLA4, are in a steady state,
representing the disease state. These T-LGL attractors are given in
the second column of Table 2, which presents the predicted T-
LGL states of 54 components of the network (all but the six source
nodes whose state is indicated at the beginning of the Results
section). We note that the two T-LGL attractors essentially
represent the same disease state since they only differ in the state of
the virtual node P2. Moreover, this disease state can be considered
as a fixed point since only two nodes oscillate in the T-LGL
attractors. For this reason we will refer to this state as the T-LGL
fixed point. It is expected that the basins of attraction of the fixed
points have similar features as those of the simplified networks.
Experimental validation of the T-LGL steady state
Experimental evidence exists for the deregulated states of 36
(67%) components out of the 54 predicted T-LGL states as
summarized in the third column of Table 2. For example, the
stable ON state of MEK, ERK, JAK, and STAT3 indicates that
the MAPK and JAK-STAT pathways are activated. The OFF
state of BID is corroborated by recent evidence that it is down-
regulated both in natural killer (NK) and in T cell LGL leukemia
[45]. In addition, the node RAS was found to be constitutively
active in NK-LGL leukemia [41], which indirectly supports our
result on the predicted ON state of this node. For three other
components, namely, GPCR, DISC, and IFNG, which were
classified as being deregulated without clear evidence of either up-
regulation or down-regulation in [18], we found that they
eventually stabilize at ON, OFF, and OFF, respectively. The
OFF
state
of
IFNG
and
DISC
is
indeed
supported
by
experimental evidence [44,46]. In the second column of Table 2,
we indicated with an asterisk the stabilized state of 17 components
that were experimentally undocumented before and thus are
predictions of our steady state analysis (P2 was not included as it is
a virtual node). We note that ten of these cases were also predicted
in [18] by simulations.
The predicted T-LGL states of these 17 components can guide
targeted experimental follow-up studies. As an example of this
approach, we tested the predicted over-activity of the node SMAD
(see Materials and Methods). As described in [18] the SMAD node
represents a merger of SMAD family members Smad 2, 3, and 4.
Smad 2 and 3 are receptor-regulated signaling proteins which are
phosphorylated and activated by type I receptor kinases while
Smad4 is an unregulated co-mediator [47]. Phosphorylated
Smad2 and/or Smad3 form heterotrimeric complexes with Smad4
and these complexes translocate to the nucleus and regulate gene
expression. Thus an ON state of SMAD in the model is a
representation of the predominance of phosphorylated Smad2
and/or phosphorylated Smad3 in T-LGL cells. In relative terms as
compared to normal (resting or activated) T cells, the predicted
ON state implies a higher level of phosphorylated Smad2/3 in T-
LGL cells as compared to normal T cells. Indeed, as shown in
Figure 6, T cells of T-LGL patients tend to have high levels of
phosphorylated Smad2/3, while normal activated T cells have
essentially no phosphorylated Smad2/3. Thus our experiments
validate the theoretical prediction.
Node perturbations
A
question
of
immense
biological
importance
is
which
manipulations of the T-LGL network can result in consistent
activation-induced cell death and the elimination of the dysreg-
ulated (diseased) behavior. We can rephrase and specify this
question as which node perturbations (knockouts or constitutive
activations) lead to a system that has only the normal fixed point.
These
perturbations
can
serve
as
candidates
for
potential
therapeutic interventions. To this end, we performed node
perturbation analysis using both structural and dynamic methods.
Structural
perturbation
analysis.
For
the
structural
analysis, using the T-LGL network (Figure 1) and the Boolean
rules (Table S1), we constructed an expanded T-LGL survival
signaling network (see Materials and Methods) as represented in
Figure 3. The state transition graph corresponding to the two
oscillatory nodes, CTLA4 and TCR. In this graph the left binary digit
of the node identifier indicates the state of CTLA4 and the right digit
represents the state of TCR. The directed edges represent state
transitions allowed by updating a single node’s state; self-loops appear
when a node is updated but its state does not change.
doi:10.1371/journal.pcbi.1002267.g003
Table 1. Boolean rules governing the nodes’ states in the 6-
node sub-network represented in Figure 2B.
Node
Boolean rule
S1P
S1P* = NOT (Ceramide OR Apoptosis)
FLIP
FLIP* = NOT (DISC OR Apoptosis)
Fas
Fas* = NOT (S1P OR Apoptosis)
Ceramide
Ceramide* = Fas AND NOT (S1P OR Apoptosis)
DISC
DISC* = (Ceramide OR (Fas AND NOT FLIP)) AND NOT Apoptosis
Apoptosis
Apoptosis* = DISC OR Apoptosis
For simplicity, the nodes’ states are represented by the node names. The
symbol * indicates the future state of the marked node.
doi:10.1371/journal.pcbi.1002267.t001
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
7
November 2011 | Volume 7 | Issue 11 | e1002267
Figure S1. In order to evaluate the importance of signaling
components mediating T-LGL leukemia, we introduced the
complementary node of Apoptosis (denoted by ,Apoptosis in
Figure S1) as an output representing the survival of the CTL cells,
which is activated by the complementary node of Caspase
(denoted by ,Caspase in Figure S1). The reason is that we are
interested in the question of how to make this outcome (i.e., the
disease state) disappear, or in graph terminology, disconnected
from the inputs of the network. In order to count all the simple
paths from a single (rather than multiple) input signal to the output
node, we fixed the states of Stimuli and IL15 at ON and those of
Stimuli2, CD45, and TAX at OFF. Once the Boolean rules were
simplified, we determined all the signaling paths from PDGF to
the output node ,Apoptosis. Interestingly, we found that the
number of signaling paths from PDGF to ,Apoptosis is much
smaller than the number of signaling paths from PDGF to
Figure 4. The state transition graph of the 6-node sub-network represented in Figure 2B. It contains 64 states of which the state shown
with a dark blue symbol is the normal fixed point and the state shown in red is the T-LGL fixed point. States denoted by light blue symbols are
uniquely in the basin of attraction of the normal fixed point whereas the states in pink can only reach the T-LGL fixed point. Gray states, on the other
hand, can lead to either fixed point.
doi:10.1371/journal.pcbi.1002267.g004
Figure 5. The probabilities of reaching the normal and T-LGL fixed points when both are reachable. These probabilities are computed
starting from the states that are shared by both basins of attraction (see gray-colored states illustrated in Figure 4).
doi:10.1371/journal.pcbi.1002267.g005
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
8
November 2011 | Volume 7 | Issue 11 | e1002267
Table 2. A summary of the dynamic analysis results of the T-LGL survival signaling network.
Node
T-LGL state
Ref.
Fixed point the disruption
leads to
Size of exclusive basin of
normal fixed point
Ref.
DISC
OFF
[46]
Normal
100%
[46]
Ceramide
OFF
[48]
Normal
100%
[48]
Caspase
OFF
[46]
Normal
100%
SPHK1
ON
[21]
Normal
100%
[18]
S1P
ON
[21]
Normal
100%
[21]
PDGFR
ON
[59]
Normal
100%
[18]
GAP
OFF*
Normal
100%
RAS
ON*
Normal
100%
[41]1
MEK
ON
[59]
Normal
100%
[41]1
ERK
ON
[50,59]
Normal
100%
[41]1
IL2RBT
ON
[60]
Normal
100%
IL2RB
ON
[60]
Normal
100%
STAT3
ON
[49]
Normal
100%
[49]
BID
OFF
[45]
Normal
100%
MCL1
ON
[49]
Normal
100%
[49]
SOCS
OFF*
Both
81%
JAK
ON
[49]
Both
81%
[49]
PI3K
ON
[50]
Both
75%
[50]
NFkB
ON
[18]
Both
75%
[18]
Fas
OFF
[48]
Both
72%
sFas
ON
[61]
Both
72%
TBET
ON
[18]
Both
63%
RANTES
ON
[44]
Both
63%
PLCG1
ON*
Both
63%
FLIP
ON
[46]
Both
56%
IL2
OFF
[62]
Both
56%
IAP
ON*
Both
56%
TNF
ON*
Both
56%
BclxL
OFF
[49]
Both
56%
GZMB
ON
[63]
Both
56%
IL2RA
OFF
[62]
Both
56%
NFAT
ON*
Both
56%
GRB2
ON*
Both
56%
IFNGT
ON
[44,62]
Both
56%
TRADD
OFF*
Both
56%
ZAP70
OFF*
Both
56%
LCK
ON
[50]
Both
56%
FYN
ON*
Both
56%
IFNG
OFF
[44]
Both
56%
SMAD
ON*
This study
Both
56%
GPCR
ON
[21,64]
Both
56%
TPL2
ON
[65]
Both
56%
A20
ON
[21]
Both
56%
IL2RAT
OFF
[62]
Both
56%
CREB
OFF*
Both
56%
P27
ON*
Both
56%
P2
ON/OFF
Both
56%
FasT
ON
[48]
T-LGL
0%
FasL
ON
[48]
T-LGL
0%
Cytoskeleton signaling
ON*
—
—
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
9
November 2011 | Volume 7 | Issue 11 | e1002267
Apoptosis (78,827 versus 346,974), consistent with the finding from
dynamic analysis that the exclusive basin of attraction of the T-
LGL fixed point is much smaller than that of the normal fixed
point.
Our goal of identifying node state manipulations that lead to the
apoptosis of the abnormally surviving T-LGL cells can be
translated into the graph-theoretical problem of finding key nodes
that mediate paths to the node ,Apoptosis. Elimination of these
nodes has the potential to make ,Apoptosis unreachable, or in
other words to make Apoptosis the only reachable outcome. The
T-LGL fixed point determined in dynamic analysis serves as a list
of candidate deletions. Accordingly, we separately deleted each
node that stabilizes at ON in the T-LGL fixed point, and each
complementary node whose corresponding original node stabilizes
at OFF in the T-LGL fixed point (see Table 2 for the state of nodes
in the T-LGL fixed point). We then calculated the importance
values of these nodes by examining the cascading effects of their
deletion on the number of simple paths from PDGF to the
,Apoptosis output (see Materials and Methods). The importance
values of the signaling components are given in Figure 7. As we
can see in this figure, several components, including ,DISC,
,Ceramide, ,Caspase, SPHK1, S1P, PDGFR, PI3K, ,SOCS,
JAK, ,GAP, RAS, NFkB, MEK, and ERK have importance
values of one (or very close to one). This means that blocking any
of these nodes disrupts (almost) all signaling paths from the source
node to ,Apoptosis, thus these nodes are candidate therapeutic
targets.
Dynamic perturbation analysis.
To identify manipulations
of the T-LGL network leading to the existence of only the normal
fixed point, we first considered the following scenario. We assumed
that the T-LGL network is the simplified network given in
Figure 2B. We examined the following dynamic perturbation
approaches as potential interventions propelling the system into
the normal fixed point. In the first two approaches, it is assumed
that the T-LGL fixed point has been already reached (i.e. the
disease has already developed), and in the last approach, all
possible initial conditions are considered.
1. Reverse the state of one node at a time in the T-LGL fixed
point for only the first time step, and keep updating the system.
This intervention may be accomplished by a pharmacological
intervention on a T-LGL cell.
2. Reverse the state of one node in the T-LGL fixed point
permanently and continue updating other nodes. This
Figure 6. Experimental validation of the increased activity (ON state) of Smad2/3 in leukemic T-LGL. Western blot detection of
phosphorylated Smad2 or Smad3, and total Smad2 (i.e. the sum of phosphorylated and non-phosphorylated Smad2) or Smad3 in activated normal T
cells compared with peripheral blood mononuclear cells from T-LGL leukemia patients confirms that Smad2 or Smad3 is unphosphorylated (inactive)
in normal T cells and predominantly phosphorylated (active) in T-LGL cells.
doi:10.1371/journal.pcbi.1002267.g006
Node
T-LGL state
Ref.
Fixed point the disruption
leads to
Size of exclusive basin of
normal fixed point
Ref.
Proliferation
OFF
[43]
—
—
Apoptosis
OFF
[66]
—
—
TCR
Oscillate*
—
—
CTLA4
Oscillate*
—
—
The first two columns from the left list the components of the network (except for the six source nodes) and their T-LGL states. The nodes’ states marked with a
* symbol were not documented experimentally in T-LGL before and were predicted by our steady state analysis. The references for the nodes’ states documented
before are given in the third column. The fixed point(s) obtained after each of the nodes’ states is reversed is given in the fourth column, while the size of the exclusive
basin of attraction of the normal fixed point, expressed as a percentage of the whole relevant state space, is indicated in the fifth column. The reference of the
perturbation cases for which experimental evidence exists is given in the last column. The first 19 nodes in the first column are potential therapeutic targets for T-LGL
leukemia.
1Evidence in NK-LGL leukemia.
doi:10.1371/journal.pcbi.1002267.t002
Table 2. Cont.
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
10
November 2011 | Volume 7 | Issue 11 | e1002267
intervention may be accomplished by genetic engineering of a
T-LGL cell.
3. Considering all possible initial states, fix the state of one node in
the opposite of its T-LGL state and keep updating other nodes.
This intervention may be accomplished by genetic engineering
of a population of CTLs.
For the first perturbation approach, we found that only the
trivial case of flipping the state of Apoptosis to ON leads
exclusively
to
the
normal
fixed
point.
Using
the
second
perturbation approach, we observed that fixing S1P at OFF or
Apoptosis at ON eliminates the T-LGL fixed point. In addition,
fixing either Ceramide or DISC at ON results in a new fixed point
which is similar to the normal fixed point of the unperturbed
system, with the only difference that the disrupted node’s state is
fixed at ON as long as the cell is alive. Using the last perturbation
approach, we found a result identical to that of the second
approach, indicating that the nodes S1P, Ceramide, and DISC are
candidate therapeutic targets for the simplified sub-network.
Experiments also confirm that Ceramide and DISC can serve as
therapeutic targets [46,48]. We note that the third approach is
superior to the second in that it provides additional information on
the size of the basin of attraction of each fixed point. For example,
we observed that in the case of over-expression of Fas, the
exclusive basin of attraction of the normal fixed point increases
significantly to 72% of the states. This suggests that although both
fixed points are still reachable, the normal fixed point is more
probable to be reached. This analysis revealed that the last
approach leads to more detailed results than the first two
approaches.
Next we focused our attention to the effects of node disruptions
on the whole network to make biologically testable predictions
about the occurrence of the disease state under different
conditions. To this end, we followed the third approach delineated
above. More precisely, for each node disruption we fixed the state
of that node in the opposite of its stabilized state in the T-LGL
fixed point given in Table 2 (i.e. we knocked out the nodes that
stabilize in the ON state in T-LGL fixed point and over-expressed
the ones that stabilize in the OFF state) and considered all possible
initial states for the remaining nodes (except for the six source
nodes). Of the 60 nodes of the network, six are source nodes, three
are output nodes and two (CTLA4 and TCR) have oscillatory
behavior in the T-LGL attractor. For each of the remaining nodes,
we fixed the state of that node in the opposite of its T-LGL state,
initiated the six source nodes as in the unperturbed case, and
identified the stabilized nodes using logical steady state analysis
(see Materials and Methods). We then simplified the network of
non-stabilized nodes according to the second step of our reduction
method (see Materials and Methods) and obtained all possible
fixed points by solving the corresponding set of Boolean equations.
For some cases we needed to construct the full state transition
graphs because of the possibility of oscillation (e.g. when the two
oscillatory nodes, CTLA4 and TCR, were connected to other
nodes in the simplified network and there was a possibility of
propagating the oscillation to other nodes in the T-LGL state). We
found that in the case of perturbation of TBET, PI3K, NFkB,
JAK, or SOCS, five additional nodes of the network connected to
CTLA4 and TCR, namely LCK, FYN, Cytoskeleton signaling,
ZAP70, and GRB2, oscillate as well. Also, for the knockout of
FYN, only two of these additional nodes, i.e. LCK and ZAP70
oscillate. In addition, in the case of perturbation of TBET, JAK,
SOCS, or IL2, the node IL2RA shows oscillatory behavior in the
T-LGL state.
In general, two types of fixed points were observed, the normal
fixed point with Apoptosis being ON and all other nodes being
OFF, and similar-to-TLGL fixed points with Apoptosis being OFF
and the state of some nodes being different from the wild-type T-
LGL fixed point due to the disruption imposed on the network.
We still consider these latter fixed points as the T-LGL fixed point.
A summary of the node disruption results, including the fixed
point(s) obtained after the disruption as well as the size of the
exclusive basin of attraction of the normal fixed point in the
respective reduced model, is given in the fourth and fifth columns
of Table 2. Our results indicate that disruption of any of the first
15 nodes in Table 2 leads to the disappearance of the T-LGL fixed
point (i.e., of the disease state). These nodes are thus predicted
candidate therapeutic targets. For example, our results suggest that
knockout of STAT3 or over-expression of Ceramide in deregu-
lated CTLs restores their activation induced cell death. We found
for the knockout of either FasT or FasL that the normal fixed point
and the 50% of the state transition graph which includes the ON
state of Apoptosis is separated from the rest of the state space and
thus they are not accessible from the biologically relevant initial
conditions. Therefore, the T-LGL fixed point is the only
biologically relevant outcome in this case. For this reason, the
size of the basin of attraction of the normal fixed point was
indicated as 0% in Table 2. Notably, these nodes can serve as
candidates for engineering of long-lived T cells, which are
Figure 7. Importance values of network components in T-LGL leukemia. These values are based on the relative reduction of the number of
paths from PDGF to ,Apoptosis after considering the cascading effects of node disruptions. The complementary nodes are denoted by the
corresponding original nodes with a symbol ‘,’ as prefix representing ‘negation’.
doi:10.1371/journal.pcbi.1002267.g007
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
11
November 2011 | Volume 7 | Issue 11 | e1002267
necessary for the delivery of virus and cancer vaccines. The
remaining node disruptions still retain both disease and normal
fixed points.
There is corroborating literature evidence for several of the
therapeutic targets predicted by our analysis. For example, it was
found experimentally that STAT3 knockdown by using siRNA or
down-regulation of MCL1 through inhibiting STAT3 induces
apoptosis in leukemic T-LGL [49]. Furthermore, in vitro Ceramide
treatment induces apoptosis in leukemic T-LGL [48]. It was also
found that treatment with IL2 and TCR stimulation facilitates
Fas-mediated apoptosis via induction of DISC formation [46]. In
addition, SPHK1 inhibition by using chemical inhibitors signifi-
cantly
induces
apoptosis
in
leukemic
T-LGL
[18].
These
experimental results validate that perturbation of these nodes
results in the normal fixed point as mentioned in Table 2.
Moreover, it was reported in [41] that inhibition of RAS through
introducing a dominant negative form of RAS, or inhibition of
MEK or ERK through chemical inhibitors, induces apoptosis in
leukemic NK-LGL, which indirectly supports our results on these
three nodes.
For the cases where both fixed points are still reachable, our
analysis of the relative size of the basins of attraction (i.e.
percentage of the whole relevant state space) of the fixed points
and the probabilities of reaching the fixed points (see Materials
and Methods) indicated that in most of these cases the trends are
similar to the wild-type model, e.g. the size of the exclusive basin of
attraction of the normal fixed point is 56%, the same as that for
the unperturbed system. In a few cases, however, including JAK,
PI3K, or NFkB knockout as well as SOCS over-expression, the
exclusive basin of attraction of the normal fixed point increased
significantly (to 75% or more). Thus, these nodes can be also
considered as potential therapeutic targets. Interestingly, for three
cases, namely JAK, PI3K, and NFkB, experimental data also
suggest that the balance between the incidence of the two fixed
points is shifted in the manipulated system compared to the
original one. For example, inhibition of JAK [49], PI3K [50] or
NFkB [18] through chemical inhibitors induces apoptosis in
leukemic T-LGL. In summary, our analysis leads to the novel
predictions that Caspase, GAP, BID, or SOCS over-expression as
well as RAS, MEK, ERK, IL2RBT, or IL2RB knockout can lead
to apoptosis of T-LGL cells.
Comparison between structural and dynamic pertur-
bation analysis.
We performed the perturbation analysis
using a dynamic method as well as a structural method. How do
the results compare? From the dynamic analysis, a node is
classified as an important mediator of the T-LGL fixed point if
reversing its state from the value it achieves in the T-LGL fixed
point will lead the system to have only the normal fixed point.
From the structural analysis, a node can be classified as an
important mediator of the T-LGL behavior if its importance value
(see Materials and Methods) to the ,Apoptosis outcome is higher
than a pre-specified threshold. We used different importance
values
as
thresholds
and
compared
the
structure-based
classification with the dynamics-based classification by using the
latter as the standard. The sensitivity (the fraction of important
components based on dynamic perturbation analysis that are
recognized as important by the structural method) and specificity
(the fraction of non-important components based on dynamic
perturbation analysis that are recognized as non-important by the
structural method) values of the structure-based classification are
summarized by the red curve in Figure 8. The structural method
gives the best fit to the dynamic method (namely, sensitivity of 1.00
and specificity of 0.76) if a threshold of 0.9 is used. An important
feature of the structural method is its incorporation of the
cascading effects of a node’s deletion. To illustrate this point, we
also show the corresponding result without considering the
cascading effects of nodes’ deletions represented by the green
curve in Figure 8. As this figure demonstrates, the results using a
pure topological measure without considering the cascading effects
gives a much worse fit to the results of the dynamic method.
Interestingly, for all the components whose manipulation lead
the system to have only the normal fixed point according to the
dynamic analysis (the first 15 components in Table 2), the reported
importance values based on the structural method were larger
than 0.95. For four additional cases, namely, SOCS, JAK, PI3K,
and NFkB, which are identified as important for survival based on
the simple path measure, the dynamic analysis results also revealed
that the T-LGL outcome has a lesser probability to be reached as
mentioned earlier. Therefore, they can also be considered as
potential therapeutic targets.
We note that there are four cases, namely, TBET, FLIP, IAP,
and TNF, which were identified as important based on the
structural method while their disruption maintains the existence of
both fixed points based on dynamic analysis and the size of the
exclusive basin of attraction of the normal fixed point is either
close to or the same as that of the wild-type system. This may be
partly due to the fact that in the state space analysis we consider all
possible initial conditions for the system, whereas the topological
analysis implicitly refers to only one initial condition, wherein
three source nodes are ON and all other nodes are OFF. Another
potential reason regarding the discrepancies between the structural
and dynamic perturbation results might be related to the structural
method’s use of the simple path measure rather than the
elementary signaling modes (ESMs, see Materials and Methods).
Furthermore, although the reduction method used for the
dynamic analysis preserves the fixed points, it can change the
state transition graph and thus may have an impact on the relative
size of the basins of attraction, serving as an alternative source of
inconsistencies. However, this change is not expected to be drastic
as we found that the exclusive basin of attraction of the normal
fixed point in the 6-node network was approximately of the same
relative size as that in the 18-node network.
Figure 8. Comparison of structural perturbation analysis
results with and without cascading effects of node deletions.
SP+CE represents the simple path measure considering cascading
effects of node deletions, and SP-CE represents the simple path
measure without considering cascading effects of node deletions.
doi:10.1371/journal.pcbi.1002267.g008
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
12
November 2011 | Volume 7 | Issue 11 | e1002267
Discussion
In this paper we presented a comprehensive analysis of the T-
LGL survival signaling network to unravel the unknown facets of
this disease. By using a reduction technique, we first identified the
fixed points of the system, namely the normal and T-LGL fixed
points, which represent the healthy and disease states, respectively.
This analysis identified the T-LGL states of 54 components of the
network, out of which 36 (67%) are corroborated by previous
experimental evidence and the rest are novel predictions. These
new predictions include RAS, PLCG1, IAP, TNF, NFAT, GRB2,
FYN, SMAD, P27, and Cytoskeleton signaling, which are
predicted to stabilize at ON in T-LGL leukemia and GAP,
SOCS, TRADD, ZAP70, and CREB which are predicted to
stabilize at OFF. In addition, we found that the node P2 can
stabilize in either the ON or OFF state, whereas two nodes, TCR
and CTLA4, oscillate. We have experimentally validated the
prediction that the node SMAD is over-active in leukemic T-LGL
by demonstrating the predominant phosphorylation of the SMAD
family members Smad2 and Smad3. The predicted T-LGL states
of other nodes provide valuable guidance for targeted experimen-
tal follow-up studies of T-LGL leukemia.
Among the predicted states, the ON state of Cytoskeleton
signaling may not be biologically relevant as this node represents
the ability of T cells to attach and move which is expected to be
reduced in leukemic T-LGL compared to normal T cells. This
discrepancy may be due to the fact that the network contains
insufficient detail regarding the regulation of the cytoskeleton, as
there is only one node, FYN, upstream of Cytoskeleton signaling
in the network. While the network is able to successfully capture
survival signaling without necessarily capturing the cytoskeleton
signaling, this discrepancy suggests that follow-up experimental
studies should be conducted to determine the relationship between
cytoskeleton signaling and survival signaling in the T-LGL
network. We note that in the case of perturbation of TBET,
PI3K, NFkB, JAK, or SOCS, the node Cytoskeleton signaling
exhibits oscillatory behavior induced by oscillations in TCR. At
present it is not known whether this predicted behavior is relevant.
Using
the
general
asynchronous
(GA)
Boolean
dynamic
approach, we analyzed the basins of attraction of the fixed points.
We found that the basin of attraction of the normal fixed point is
larger than that of the T-LGL fixed point. The trajectories starting
from each initial state toward the T-LGL fixed point (Figure 4)
may be indicative of the accumulating deregulations that lead to
the disease-associated stable survival state. Although the fixed
points, being time independent, are the same for all update
methods or implementations of time, the update method may
affect the structure of the state transition graph of the system and
the basins of attraction of the fixed points. We note that the GA
method assumes that each node has an equal chance of being
updated. If quantitative or kinetic information becomes available
in this system, unequal probabilities may be implemented by
grouping the nodes into several ‘‘priority classes’’ and assigning a
weight to each class where higher weights indicate more probable
transitions [51]. Incorporating such information into the state
space may prune the allowed trajectories and give further insights
into the accumulation of deregulations.
We took one step further by performing a perturbation analysis
using dynamical and structural methods to identify the interven-
tions leading to the disappearance of the disease fixed point. We
note that our study has a dramatically larger scope than the
previous key mediator analysis of Zhang et al [18]. For the
dynamical analysis, we employed the GA approach instead of the
random order asynchronous method and considered all possible
initial conditions as opposed to performing numerical simulations
using a specific initial condition. Zhang et al only focused on the
node Apoptosis, and identified as ‘‘key mediators’’ the nodes
whose altered state increases the frequency of ON state of
Apoptosis. An increase in Apoptosis’ ON state does not necessarily
imply that apoptosis is the only possible final outcome of the
system. In this work, after finding the fixed points, which
completely describe the state of the whole system, we performed
dynamic perturbation analysis by fixing the state of each node to
its opposite state in the T-LGL fixed point and determining which
fixed points were obtained and what their basins of attraction
were. This way we were able to identify and distinguish the key
mediators whose altered state completely eliminates the leukemic
outcome, and those whose altered state reduces the basin of
attraction
of
the
leukemic
outcome.
Moreover,
numerical
simulations, as done in [18], may not be able to thoroughly
sample different timing. In this study, using a reduction technique,
we found the cases when timing does not matter with certainty
(where there is only one fixed point), and also the cases in which
timing and initial conditions may matter (where there are two
reachable fixed points). For the perturbation analysis using the
structural method, we used the simple path (SP) measure to
identify important mediators of the disease outcome and observed
consistent results with the dynamic analysis. Our dynamical and
structural analysis led to the identification of 19 therapeutic targets
(the first 19 nodes in the first column of Table 2), 53% of which are
supported by direct experimental evidence and 15% of which are
supported by indirect evidence.
Multi-stability (having multiple steady states) is an intrinsic
dynamic property of many disease networks [52,53], which is
related to the presence of feedback loops in the network. In a
graph-theoretical sense, a feedback loop is a directed cycle whose
sign depends upon the parity of the number of negative
interactions in the cycle. A positive/negative feedback loop has
an even/odd number of negative interactions. It was conjectured
that the presence of positive feedback loops in the network is
necessary for multi-stability whereas the existence of negative
feedback loops is required for having sustained oscillations [54].
From a biological point of view, the former dynamical property is
associated with multiple cell types after differentiation while the
latter is related to stable periodic behaviors such as circadian
rhythms [55]. We note that the T-LGL signaling network consists
of both positive and negative feedbacks and thus has a potential for
both multi-stability and oscillations. Indeed, the negative feedback
in the top sub-graph of Figure 2A causes the complex attractor
shown in Figure 3. In contrast, the negative feedback on the node
P2 of the bottom sub-graph is counteracted by the positive self-
loop on the same node, thus no complex attractor is possible for
the bottom sub-graph of Figure 2A. The two mutual inhibition-
type positive feedback loops present in the bottom sub-graph and
the self-loop on P2 generate the three fixed points, while the
positive self-loop on Apoptosis maintains the normal fixed point
once Apoptosis is turned ON.
Negative feedback loops can be a source of oscillations [56],
homeostasis [56], or excitation-adaptation behavior [57]. Espe-
cially, when the activation is slower than the inhibitory interaction
in the negative feedback, it can lead to sustained oscillations [56].
In the T-LGL network, the negative feedback loop between the T
cell receptor TCR and CTLA4 modulates stimulus-induced
activation of the receptor in such a way that CTLA4 is indirectly
activated after prolonged TCR activation, whereas the inhibition
of TCR by CTLA4 is a direct interaction [58]. That is, activation
is slower than inhibition in the negative feedback and thus an
oscillatory behavior reminiscent of that obtained by our asyn-
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
13
November 2011 | Volume 7 | Issue 11 | e1002267
chronous Boolean model would also be observed in continuous
modeling frameworks as well. Although no time-measurements of
the T cell receptor activity in T-LGL exist, it has been reported
that there is variability for TCR activation in different patients
([43] and unpublished observation by T.P. Loughran), supporting
the absence of a steady state behavior.
Our study revealed that both structural and dynamic analysis
methods can be employed to identify therapeutic targets of a
disease, however, they differ in implementation efficiency as well
as the scope and applicability of the results. The structural analysis
does not require mapping of the state space and thus is less
computationally intensive and is more feasible for large network
analysis, but it may not capture all the initial states and thus may
miss or inaccurately identify some important features. The
dynamic analysis method, while computationally intensive, yields
a comprehensive picture of the state transition graph, including all
possible fixed points of the system, their corresponding basins of
attraction, as well as the relative frequency of trajectories leading
to each fixed point. We demonstrated that the limitations related
to the vast state space of large networks can be overcome by
judicious use of the network reduction technique that we
developed in our previous study [22]. We conclude that the
structural method incorporating the cascading effects of node
disruptions is best employed for quick exploratory analysis, and
dynamic analysis should be performed to get a thorough and
detailed insight into the behavior of a system. Overall, the
combined analysis presented in this study opens a promising
avenue to predict dysregulated components and identify potential
therapeutic targets, and it is versatile enough to be successfully
applied to a large variety of signal transduction and regulatory
networks related to diseases.
Supporting Information
Figure
S1
The
expanded
T-LGL
survival
signaling
network. Composite nodes are represented by small gray solid
circles, original nodes are represented by large ovals, and
complementary nodes are represented by rectangles. The labels
of complementary nodes are denoted by the labels for the
corresponding original nodes with a symbol ‘,’ as prefix
representing ‘negation’.
(TIF)
Table S1
Boolean rules governing the state of the T-LGL
signaling network depicted in Figure 1. For simplicity, the
nodes’ states are represented by the node names. The symbol
* indicates the future state of the marked node. The Boolean rule
for each node is determined based on the nature of interactions
between that node and the nodes directly interacting with it. This
rule can be expressed using the logical operators AND, OR and
NOT. For example, if the given node has a single upstream node,
the corresponding Boolean function would include only one
variable. This variable will be combined with a NOT operator if
the upstream node is an inhibitor. In cases where the given node
has multiple upstream nodes, their effect is combined with AND or
OR operators (potentially in conjunction with the NOT operator)
to correctly recast the regulatory interactions. For example, the
AND operator is used when the co-expression of two (or more)
activating inputs is required for activating the target node,
whereas, the OR operator implies that the activity of at least
one of the upstream activators is sufficient to activate the target
node. The type of each interaction (i.e. the logical rule) should be
extracted from the relevant literature and experimental evidence.
This table is adapted from [1]. The interested reader is referred to
[1] for the detailed explanation of the rules.
(PDF)
Table S2
The full names of components in the T-LGL
signaling network corresponding to the abbreviated
node labels used in Figure 1. Several network nodes represent
the union of a few proteins with similar roles. In such cases, a
single entry in the first column corresponds to several entries in the
second column. This table and its caption are adapted from [1].
(PDF)
Table S3
Boolean rules governing the state of the 18-
node sub-network depicted in Figure 2A. For simplicity, the
nodes’ states are represented by the node names. The symbol
* indicates the future state of the marked node.
(PDF)
Acknowledgments
The authors would like to thank Dr. Ranran Zhang for fruitful discussions.
Author Contributions
Conceived and designed the experiments: RA AS RSW XL TPL.
Performed the experiments: AS RSW AL. Analyzed the data: AS RSW
XL TPL RA. Contributed reagents/materials/analysis tools: AS RSW IA.
Wrote the paper: AS RA.
References
1. Abdi A, Tahoori MB, Emamian ES (2008) Fault diagnosis engineering of digital
circuits can identify vulnerable molecules in complex cellular pathways. Sci
Signal 1: ra10.
2. Leibiger IB, Brismar K, Berggren PO (2010) Novel aspects on pancreatic beta-
cell signal-transduction. Biochem Biophys Res Commun 396: 111–115.
3. Muscogiuri G, Chavez AO, Gastaldelli A, Perego L, Tripathy D, et al. (2008)
The crosstalk between insulin and renin-angiotensin-aldosterone signaling
systems and its effect on glucose metabolism and diabetes prevention. Curr
Vasc Pharmacol 6: 301–312.
4. Gordon KJ, Blobe GC (2008) Role of transforming growth factor-b superfamily
signaling pathways in human disease. Biochim Biophys Acta 1782: 197–228.
5. Mavers M, Ruderman EM, Perlman H (2009) Intracellular signal pathways:
potential for therapies. Curr Rheumatol Rep 11: 378–385.
6. Ikushima H, Miyazono K (2010) TGFb signalling: a complex web in cancer
progression. Nat Rev Cancer 10: 415–424.
7. Sokol L, Loughran TP, Jr. (2006) Large granular lymphocyte leukemia.
Oncologist 11: 263–273.
8. Shah MV, Zhang R, Loughran TP, Jr. (2009) Never say die: survival signaling in
large granular lymphocyte leukemia. Clin Lymphoma Myeloma 9 Suppl 3:
S244–253.
9. Albert R, Barabasi AL (2002) Statistical mechanics of complex networks. Rev
Mod Phys 74: 47–97.
10. Lee TI, Rinaldi NJ, Robert F, Odom DT, Bar-Joseph Z, et al. (2002)
Transcriptional regulatory networks in Saccharomyces cerevisiae. Science 298:
799–804.
11. Li S, Armstrong CM, Bertin N, Ge H, Milstein S, et al. (2004) A map of the
interactome network of the metazoan C. elegans. Science 303: 540–543.
12. Kervizic G, Corcos L (2008) Dynamical modeling of the cholesterol regulatory
pathway with Boolean networks. BMC Syst Biol 2: 99.
13. Li S, Assmann SM, Albert R (2006) Predicting essential components of signal
transduction networks: a dynamic model of guard cell abscisic acid signaling.
PLoS Biol 4: e312.
14. Saez-Rodriguez J, Simeoni L, Lindquist JA, Hemenway R, Bommhardt U, et al.
(2007) A logical model provides insights into T cell receptor signaling. PLoS
Comput Biol 3: e163.
15. Albert R, Wang RS (2009) Discrete dynamic modeling of cellular signaling
networks. Methods Enzymol 467: 281–306.
16. Kauffman SA (1969) Metabolic stability and epigenesis in randomly constructed
genetic nets. J Theor Biol 22: 437–467.
17. Thomas R (1973) Boolean formalization of genetic control circuits. J Theor Biol
42: 563–585.
18. Zhang R, Shah MV, Yang J, Nyland SB, Liu X, et al. (2008) Network model of
survival signaling in large granular lymphocyte leukemia. Proc Natl Acad
Sci U S A 105: 16308–16313.
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
14
November 2011 | Volume 7 | Issue 11 | e1002267
19. Layek R, Datta A, Bittner M, Dougherty ER (2011) Cancer therapy design
based on pathway logic. Bioinformatics 27: 548–555.
20. Weinberg RA (2006) The Biology of Cancer. Princeton: Garland Science.
21. Shah MV, Zhang R, Irby R, Kothapalli R, Liu X, et al. (2008) Molecular
profiling of LGL leukemia reveals role of sphingolipid signaling in survival of
cytotoxic lymphocytes. Blood 112: 770–781.
22. Saadatpour A, Albert I, Albert R (2010) Attractor analysis of asynchronous
Boolean models of signal transduction networks. J Theor Biol 266: 641–656.
23. Wang RS, Albert R (2011) Elementary signaling modes predict the essentiality of
signal transduction network components. BMC Syst Biol 5: 44.
24. Kauffman SA (1993) Origins of order: self-organization and selection in
evolution. New York: Oxford University Press.
25. Thomas R, D’Ari R (1990) Biological Feedback. Boca Raton: CRC Press.
26. Papin JA, Hunter T, Palsson BO, Subramaniam S (2005) Reconstruction of
cellular signalling networks and analysis of their properties. Nat Rev Mol Cell
Biol 6: 99–111.
27. Chaves M, Albert R, Sontag ED (2005) Robustness and fragility of Boolean
models for genetic regulatory networks. J Theor Biol 235: 431–449.
28. Chaves M, Sontag ED, Albert R (2006) Methods of robustness analysis for
Boolean models of gene control networks. Syst Biol (Stevenage) 153: 154–167.
29. Harvey I, Bossomaier T (1997) Time out of joint: Attractors in asynchronous
random Boolean networks. In: Husbands P, Harvey I, eds. Cambridge, U.K. pp
67–75.
30. Sevim V, Gong X, Socolar JE (2010) Reliability of transcriptional cycles and the
yeast cell-cycle oscillator. PLoS Comput Biol 6: e1000842.
31. Faure A, Naldi A, Chaouiya C, Thieffry D (2006) Dynamical analysis of a
generic Boolean model for the control of the mammalian cell cycle.
Bioinformatics 22: e124–131.
32. Garg A, Di Cara A, Xenarios I, Mendoza L, De Micheli G (2008) Synchronous
versus asynchronous modeling of gene regulatory networks. Bioinformatics 24:
1917–1925.
33. Albert I, Thakar J, Li S, Zhang R, Albert R (2008) Boolean network simulations
for life scientists. Source Code Biol Med 3: 16.
34. Grinstead CM, Snell JL (1997) Introduction to Probability. Providence, RI: AMS.
35. Naldi A, Remy E, Thieffry D, Chaouiya C (2011) Dynamically consistent
reduction of logical regulatory graphs. Theor Comput Sci 412: 2207–2218.
36. Veliz-Cuba A (2011) Reduction of Boolean network models. J Theor Biol 289C:
167–172.
37. Klamt S, Saez-Rodriguez J, Gilles ED (2007) Structural and functional analysis
of cellular networks with CellNetAnalyzer. BMC Syst Biol 1: 2.
38. Klamt S, Saez-Rodriguez J, Lindquist JA, Simeoni L, Gilles ED (2006) A
methodology for the structural and functional analysis of signaling and
regulatory networks. BMC Bioinformatics 7: 56.
39. Stelling J, Klamt S, Bettenbrock K, Schuster S, Gilles ED (2002) Metabolic
network structure determines key aspects of functionality and regulation. Nature
420: 190–193.
40. Palumbo MC, Colosimo A, Giuliani A, Farina L (2005) Functional essentiality
from topology features in metabolic networks: a case study in yeast. FEBS Lett
579: 4642–4646.
41. Epling-Burnette PK, Bai F, Wei S, Chaurasia P, Painter JS, et al. (2004) ERK
couples chronic survival of NK cells to constitutively activated Ras in
lymphoproliferative disease of granular lymphocytes (LDGL). Oncogene 23:
9220–9229.
42. Kachalo S, Zhang R, Sontag E, Albert R, DasGupta B (2008) NET-
SYNTHESIS: a software for synthesis, inference and simplification of signal
transduction networks. Bioinformatics 24: 293–295.
43. Aprile JA, Russo M, Pepe MS, Loughran TP, Jr. (1991) Activation signals
leading to proliferation of normal and leukemic CD3+ large granular
lymphocytes. Blood 78: 1282–1285.
44. Kothapalli R, Nyland SB, Kusmartseva I, Bailey RD, McKeown TM, et al.
(2005) Constitutive production of proinflammatory cytokines RANTES, MIP-1b
and IL-18 characterizes LGL leukemia. Int J Oncol 26: 529–535.
45. Hodge DL, Yang J, Buschman MD, Schaughency PM, Dang H, et al. (2009)
Interleukin-15 enhances proteasomal degradation of bid in normal lymphocytes:
implications for large granular lymphocyte leukemias. Cancer Res 69:
3986–3994.
46. Yang J, Epling-Burnette PK, Painter JS, Zou J, Bai F, et al. (2008) Antigen
activation and impaired Fas-induced death-inducing signaling complex
formation in T-large-granular lymphocyte leukemia. Blood 111: 1610–1616.
47. Shi Y, Massague J (2003) Mechanisms of TGF-b signaling from cell membrane
to the nucleus. Cell 113: 685–700.
48. Lamy T, Liu JH, Landowski TH, Dalton WS, Loughran TP, Jr. (1998)
Dysregulation of CD95/CD95 ligand-apoptotic pathway in CD3+ large
granular lymphocyte leukemia. Blood 92: 4771–4777.
49. Epling-Burnette PK, Liu JH, Catlett-Falcone R, Turkson J, Oshiro M, et al.
(2001) Inhibition of STAT3 signaling leads to apoptosis of leukemic large
granular lymphocytes and decreased Mcl-1 expression. J Clin Invest 107:
351–362.
50. Schade AE, Powers JJ, Wlodarski MW, Maciejewski JP (2006) Phosphatidyli-
nositol-3-phosphate kinase pathway activation protects leukemic large granular
lymphocytes from undergoing homeostatic apoptosis. Blood 107: 4834–4840.
51. Tournier L, Chaves M (2009) Uncovering operational interactions in genetic
networks using asynchronous Boolean dynamics. J Theor Biol 260: 196–209.
52. Frohlich F, Sejnowski TJ, Bazhenov M (2010) Network bistability mediates
spontaneous transitions between normal and pathological brain states. J Neurosci
30: 10734–10743.
53. Shiraishi T, Matsuyama S, Kitano H (2010) Large-scale analysis of network
bistability for human cancers. PLoS Comput Biol 6: e1000851.
54. Thomas R (1981) On the relation between the logical structure of systems and
their ability to generate multiple steady states and sustained oscillations. In:
Numerical Methods in the Study of Critical Phenomena. Berlin: Springer
Verlag. pp 180–193.
55. Thomas R, Thieffry D, Kaufman M (1995) Dynamical behaviour of biological
regulatory networks–I. Biological role of feedback loops and practical use of the
concept of the loop-characteristic state. Bull Math Biol 57: 247–276.
56. Tyson JJ, Chen KC, Novak B (2003) Sniffers, buzzers, toggles and blinkers:
dynamics of regulatory and signaling pathways in the cell. Curr Opin Cell Biol
15: 221–231.
57. Alon U (2006) An Introduction to Systems Biology: Design Principles of
Biological Circuits. Boca Raton, FL: Chapman & Hall/CRC.
58. Saito T, Yamasaki S (2003) Negative feedback of T cell activation through
inhibitory adapters and costimulatory receptors. Immunol Rev 192: 143–160.
59. Yang J, Liu X, Nyland SB, Zhang R, Ryland LK, et al. (2010) Platelet-derived
growth factor mediates survival of leukemic large granular lymphocytes via an
autocrine regulatory pathway. Blood 115: 51–60.
60. Zambello R, Facco M, Trentin L, Sancetta R, Tassinari C, et al. (1997)
Interleukin-15 triggers the proliferation and cytotoxicity of granular lymphocytes
in patients with lymphoproliferative disease of granular lymphocytes. Blood 89:
201–211.
61. Liu JH, Wei S, Lamy T, Li Y, Epling-Burnette PK, et al. (2002) Blockade of Fas-
dependent apoptosis by soluble Fas in LGL leukemia. Blood 100: 1449–1453.
62. Loughran TP, Jr., Aprile JA, Ruscetti FW (1990) Anti-CD3 monoclonal
antibody-mediated cytotoxicity occurs through an interleukin-2-independent
pathway in CD3+ large granular lymphocytes. Blood 75: 935–940.
63. Kothapalli R, Bailey RD, Kusmartseva I, Mane S, Epling-Burnette PK, et al.
(2003) Constitutive expression of cytotoxic proteases and down-regulation of
protease inhibitors in LGL leukemia. Int J Oncol 22: 33–39.
64. Kothapalli R, Kusmartseva I, Loughran TP (2002) Characterization of a human
sphingosine-1-phosphate receptor gene (S1P5) and its differential expression in
LGL leukemia. Biochim Biophys Acta 1579: 117–123.
65. Christoforidou AV, Papadaki HA, Margioris AN, Eliopoulos GD, Tsatsanis C
(2004) Expression of the Tpl2/Cot oncogene in human T-cell neoplasias. Mol
Cancer 3: 34.
66. Loughran TP, Jr. (1993) Clonal diseases of large granular lymphocytes. Blood
82: 1–14.
Dynamical and Structural Analysis of T-LGL Network
PLoS Computational Biology | www.ploscompbiol.org
15
November 2011 | Volume 7 | Issue 11 | e1002267
|
22102804
|
Caspase = ( ( ( BID ) AND NOT ( IAP ) ) AND NOT ( Apoptosis ) ) OR ( ( DISC ) AND NOT ( Apoptosis ) )
Apoptosis = ( Apoptosis ) OR ( Caspase )
sFas = ( ( S1P ) AND NOT ( Apoptosis ) )
MCL1 = NOT ( ( Apoptosis ) OR ( DISC ) )
Ceramide = ( ( ( Fas ) AND NOT ( Apoptosis ) ) AND NOT ( S1P ) )
IFNG = NOT ( ( Apoptosis ) OR ( P2 ) OR ( SMAD ) )
P2 = ( ( P2 ) AND NOT ( Apoptosis ) ) OR ( ( IFNG ) AND NOT ( Apoptosis ) )
IAP = NOT ( ( Apoptosis ) OR ( BID ) )
GPCR = ( ( S1P ) AND NOT ( Apoptosis ) )
SMAD = ( ( GPCR ) AND NOT ( Apoptosis ) )
DISC = ( ( ( Fas ) AND NOT ( FLIP ) ) AND NOT ( Apoptosis ) ) OR ( ( Ceramide ) AND NOT ( Apoptosis ) )
BID = NOT ( ( Apoptosis ) OR ( MCL1 ) )
CTLA4 = ( ( TCR ) AND NOT ( Apoptosis ) )
CREB = ( ( IFNG ) AND NOT ( Apoptosis ) )
Fas = NOT ( ( Apoptosis ) OR ( sFas ) )
S1P = NOT ( ( Apoptosis ) OR ( Ceramide ) )
FLIP = NOT ( ( Apoptosis ) OR ( DISC ) )
TCR = NOT ( ( Apoptosis ) OR ( CTLA4 ) )
|
Network Model of Immune Responses Reveals Key
Effectors to Single and Co-infection Dynamics by a
Respiratory Bacterium and a Gastrointestinal Helminth
Juilee Thakar1,2, Ashutosh K. Pathak1,3, Lisa Murphy4¤, Re´ka Albert1,5, Isabella M. Cattadori1,3*
1 Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America, 2 Department of Pathology, Yale
University School of Medicine, New Haven, Connecticut, United States of America, 3 Department of Biology, The Pennsylvania State University, University Park,
Pennsylvania, United States of America, 4 Division of Animal Production and Public Health, Veterinary School, University of Glasgow, Glasgow, United Kingdom,
5 Department of Physics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
Abstract
Co-infections alter the host immune response but how the systemic and local processes at the site of infection interact is
still unclear. The majority of studies on co-infections concentrate on one of the infecting species, an immune function or
group of cells and often focus on the initial phase of the infection. Here, we used a combination of experiments and
mathematical modelling to investigate the network of immune responses against single and co-infections with the
respiratory bacterium Bordetella bronchiseptica and the gastrointestinal helminth Trichostrongylus retortaeformis. Our goal
was to identify representative mediators and functions that could capture the essence of the host immune response as a
whole, and to assess how their relative contribution dynamically changed over time and between single and co-infected
individuals. Network-based discrete dynamic models of single infections were built using current knowledge of bacterial
and helminth immunology; the two single infection models were combined into a co-infection model that was then verified
by our empirical findings. Simulations showed that a T helper cell mediated antibody and neutrophil response led to
phagocytosis and clearance of B. bronchiseptica from the lungs. This was consistent in single and co-infection with no
significant delay induced by the helminth. In contrast, T. retortaeformis intensity decreased faster when co-infected with the
bacterium. Simulations suggested that the robust recruitment of neutrophils in the co-infection, added to the activation of
IgG and eosinophil driven reduction of larvae, which also played an important role in single infection, contributed to this
fast clearance. Perturbation analysis of the models, through the knockout of individual nodes (immune cells), identified the
cells critical to parasite persistence and clearance both in single and co-infections. Our integrated approach captured the
within-host immuno-dynamics of bacteria-helminth infection and identified key components that can be crucial for
explaining individual variability between single and co-infections in natural populations.
Citation: Thakar J, Pathak AK, Murphy L, Albert R, Cattadori IM (2012) Network Model of Immune Responses Reveals Key Effectors to Single and Co-infection
Dynamics by a Respiratory Bacterium and a Gastrointestinal Helminth. PLoS Comput Biol 8(1): e1002345. doi:10.1371/journal.pcbi.1002345
Editor: Rob J. De Boer, Utrecht University, Netherlands
Received August 3, 2011; Accepted November 25, 2011; Published January 12, 2012
Copyright: 2012 Thakar et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This study, AKP and LM were supported by a Human Frontier Science Program grant (RGP0020/2007-C). RA was supported by NSF grant CCF-0643529.
The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: imc3@psu.edu
¤ Current address: Life Technologies Ltd, Paisley, United Kingdom
Introduction
Hosts that are immunologically challenged by one infection often
show increased susceptibility to a second infectious agent, whether a
micro- or a macro-parasite. Changes in the immune status and
polarization of the response towards one parasite can indeed
facilitate the establishment and survival of a second parasitic species
[1–3]. At the level of the individual host, this can be described as an
immune system that has to optimize the specificity and effectiveness
of the responses against different infections while engaging in
secondary but equally important functions, like tissue repair or
avoiding immuno-pathology. Systemic cross-regulatory processes
and bystander effects by T helper cells (Th) maintain control of these
functions both at the systemic and local level [4–8]. Concurrent
parasite infections are regulated by and affect these mechanisms
[2,4,9–14]. They can also influence each other directly, when
sharing the same tissue [15–16] or through the immune system via
passive effects or active manipulation of the immune components, if
colonizing different organs [4,9–14].
Empirical work on bacteria-macroparasite co-infections has
often found that the development of a Th2 mediated response
towards the helminth leads to a reduction of the protective Th1
cytokine response against the bacteria and a more severe bacteria-
induced pathology [4,11–14], although a decrease of tissue
atrophy has also been observed [17–18]. The suppression of
Th1 cell proliferation acts both on the inductors and effectors and
is mainly driven by the repression of the IFNc mediated
inflammatory activity during the early stages of the infection.
However, the degree of the T helper cell polarization and the
kinetics of effectors depend on the type, intensity and duration of
the co-infection, over and above the very initial immune status of
the host. Since host immunity is both a major selective pressure for
parasite transmission and host susceptibility to re-infections, the
presence of one infection can have major consequences for the
PLoS Computational Biology | www.ploscompbiol.org
1
January 2012 | Volume 8 | Issue 1 | e1002345
spread and persistence of the second infection. For example,
Mycobacterium
tuberculosis
induces
more
severe
disease
when
concurrent with intestinal helminths, suggesting increased host
infectiousness and bacterial transmission compared to single
infected individuals [14].
Understanding how the infection by a second parasite species
can
influence
the
network
of
immune
processes
and
the
polarization towards one of the infecting agents requires the
quantification of the immune components both at the systemic
level and at the local site of infection, and the ability to follow the
kinetics of these processes over time. The immunology of co-
infection often considers the Th1/Th2 paradigm a tractable
simplification of the overall immune response and its main
functions. Yet, this approach tells us only half of the story, namely
the systemic component. Indeed, organ compartmentalization and
tissue specificity create well defined host-parasite environments
that contribute to, as well as are modulated by, the immune system
as a whole [19–20]. This brings us to the questions: what are the
key processes and components that capture the essence of immune
mediated parasite interactions in co-infections? And, how do these
differ from single infections?
To address these questions we used a combination of laboratory
experiments and network-based discrete dynamic modelling, and
examined changes in the immune response against single and co-
infection with the respiratory bacterium Bordetella bronchiseptica and
the gastrointestinal helminth Trichostrongylus retortaeformis, two
common infections of the European rabbit (Oryctolagus cuniculus).
Both parasites cause persistent infections that occur with high
prevalence and intensity in free-living rabbit populations [21–22].
B. bronchiseptica is a gram-negative bacterium that colonizes the
respiratory tract through oral-nasal transmission and usually
results in asymptomatic infections. B. bronchiseptica has been largely
isolated in wildlife, pets and livestock but rarely in humans [23]
where it is out-competed by the human-specific Bordetella pertussis
and Bordetella parapertussis, the etiological agents of whooping cough
[24]. Previous empirical and modelling work in a murine system
showed that the bacterium induces anti-inflammatory responses by
modulating Th regulation, thereby facilitating bacterial establish-
ment
and
proliferation
[8,25].
However,
hosts
successfully
counteract the pathogen mediated inhibitions by activating a
protective Th1 cell mediated IFNc response, which leads to
bacterial clearance from the lower respiratory tract, but not the
nasal cavity, via Fc receptor mediated phagocytosis [25–27]. Our
recent laboratory studies of rabbits infected with B. bronchiseptica
agree with the general findings of bacterial clearance from the
lower respiratory tract but persistence in the nasal cavity [28].
The gastrointestinal helminth T. retortaeformis has a direct life
cycle and colonizes the small intestine following ingestion of
pasture contaminated with infective third stage larvae (L3). The
majority of larvae settle in the duodenum where they develop into
adults in about 11 days [29]. A model of the seasonal dynamics of
the T. retortaeformis-rabbit interaction suggested that acquired
immunity develops proportionally to the accumulated exposure to
infection and successfully reduces helminth intensity in older hosts
[21,30]. These results were recently confirmed by challenging
laboratory rabbits with a primary infection of T. retortaeformis where
the quick production of antibodies and eosinophils was associated
with the consistent reduction but not complete clearance of the
helminth by 120 days post challenge [31].
Based on previous studies on bacteria-macroparasite co-
infections and our recent work on the rabbit system, we
hypothesized that during a B. bronchiseptica-T. retortaeformis co-
infection the presence of helminths will delay bacterial clearance
from the respiratory tract but there will be no change in helminth
abundance in the small intestine. We predicted a T. retortaeformis
mediated Th2 polarization at the systemic level and a bystander
effect in the distal respiratory tract. This will have suppressed
IFNc, resulting in the enhancement of bacterial intensity and
deferred clearance in the lower respiratory tract compared to
single infection. We also expected the Th2 systemic environment
to control helminth abundance but not to change the numbers
compared to the single infection. To examine our hypothesis,
laboratory data on single infections were used to build discrete
dynamic models describing the immune processes generated in
response to each infection. The two single infection models were
then connected through the cross-modulation of Th cells and the
cytokine network at the systemic level, and allowed to reflect
changes in these interactions at the local level. The resulting co-
infection model and the dynamics of the parasites were finally
compared with our laboratory experiment of bacteria-helminth
co-infection to confirm the correctness of the model. Lastly, we
examined the robustness of the immune networks with respect to
the deactivation of single immune nodes by simulated knockout
laboratory experiments. In other words, we tested the role of a
large number of immune components, how their knockout affected
the dynamics of infection and how the system converged into a
potentially novel stable state. This allowed us to elucidate the
immune key mechanisms and pathways behind the observed
dynamics
and
the
relative
differences
between
single
and
co-infection.
Results
The causal interactions between the immune components
activated by B. bronchiseptica and T. retortaeformis were assembled
in the form of two distinct pathogen-specific networks of immune
responses. The network of interactions against B. bronchiseptica was
based on the infection in the lungs, the crucial organ for bacterial
clearance, and constructed following Thakar et al. [8] and the
current knowledge of the dynamics of B. bronchiseptica infection in
mice (Fig. 1). There is a rich literature on the immunology of
gastrointestinal helminth infections and important general features
can be identified despite the fact that these mechanisms are often
Author Summary
Infections with different infecting agents can alter the
immune response against any one parasite and the relative
abundance and persistence of the infections within the
host. This is because the immune system is not compart-
mentalized but acts as a whole to allow the host to
maintain control of the infections as well as repair
damaged tissues and avoid immuno-pathology. There is
no comprehensive understanding of the immune respons-
es during co-infections and of how systemic and local
mechanisms interact. Here we integrated experimental
data with mathematical modelling to describe the network
of immune responses of single and co-infection by a
respiratory bacterium and a gastrointestinal helminth. We
were able to identify key cells and functions responsible
for clearing or reducing both parasites and showed that
some mechanisms differed between type of infection as a
result of different signal outputs and cells contributing to
the immune processes. This study highlights the impor-
tance of understanding the immuno-dynamics of co-
infection as a host response, how immune mechanisms
differ from single infections and how they may alter
parasite persistence, impact and abundance.
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
2
January 2012 | Volume 8 | Issue 1 | e1002345
species-specific. The immune network against T. retortaeformis was
built on the knowledge of helminth infections in mice [6–7] and
focused on the duodenum (the first section of the small intestine),
where the majority of T. retortaeformis colonization and immune
activity was observed (Fig. 2) [29,31]. Both networks were
characterized by two connected compartments: Compartment I
represented the immune interactions at the local site of infection,
the lungs or duodenum, while Compartment II described the
systemic site of T and B cell activation and differentiation, for
example, the lymph node.
The networks were then developed into discrete dynamic
models [32]. Discrete dynamic modelling has been proven to be a
feasible and useful approach to qualitatively characterize systems
where the detailed information necessary for quantitative models is
lacking [32–33]. For our purpose to examine the pattern of
immune responses to single and co-infection at the local and
systemic level and, importantly, to highlight key interactions and
cells that generated the pattern observed, the framework of the
discrete dynamic Boolean model appeared to be a robust and
tractable choice [34–36], given that the kinetics and timescales of
many of the immune interactions is unknown in the rabbit system.
Each node (e.g. immune cell) was categorized by two qualitative
states, ON and OFF, which are determined from the regulation of
the focal node by upstream nodes given in the network. This
regulation is given by a Boolean transfer function [32,34–35] (see
Materials and Methods, and Supplement Text S1). The nodes in
the ON state are assumed to be above an implicit threshold that
can be defined as the concentration necessary to activate
downstream immune processes; below this threshold the node is
in an OFF state. To follow the dynamical status of the system
through time, we repeatedly applied the Boolean transfer functions
for each node until a steady state (i.e. clearance of the pathogen)
was found. To determine the node consensus activity over time
(i.e. the time course of cell concentration or parasite numbers
Figure 1. Network of immune components considered in single B. bronchiseptica infection. Ovals represent network nodes and indicate
the node name in an abbreviated manner. Compartment I denotes the nodes in the lungs and Compartment II combines the nodes at systemic level.
Terminating black arrows on an edge indicate positive effects (activation) and terminating red blunt segments indicate negative effects (inhibition).
Grey nodes have been quantified in the single laboratory experiment. Abbreviations: Bb: B. bronchiseptica; Oag: O-antigen; IL4II: Interleukin 4 in the
systemic compartment; NE: Recruited neutrophils; IL12I: Interleukin 12 in lungs; IgA: Antibody A; C: Complement; TrII: T regulatory cells in the
systemic compartment; IL4I: Interleukin 4 in the lungs; Th2II: Th2 cells in the systemic compartment; TrI: T regulatory cells in the lungs; Th2I: Th2
cells in the lungs; IL10II: Interleukin 10 in the lymph nodes; TTSSII: Type three secretion system in the lymph nodes; TTSSI: Type three secretion
system in the lungs; IgG: Antibody G; IL10I: Interleukin 10 in the lungs; IFNcI: Interferon gamma in the lungs; IL12II: Interleukin 12 in the systemic
compartment; BC: B cells; DCII: Dendritic cells in the systemic compartment; DCI: Dendritic cells in the lungs; Th1I: T helper cell subtype I in the
lungs; PIC: Pro-inflammatory cytokines; Th1II: T helper cell subtype I in the systemic compartment EC: Epithelial cells; AP: Activated phagocytes; T0:
Naı¨ve T cells; AgAb: Antigen-antibody complexes; MP: Macrophages in the lungs; DNE: dead neutrophils; PH: Phagocytosis.
doi:10.1371/journal.pcbi.1002345.g001
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
3
January 2012 | Volume 8 | Issue 1 | e1002345
shared by multiple trajectories) we ran the simulations 100 times
by randomly sampling timescales and plotted each node activity
profile, defined as the proportion of simulations in which the node
is in the ON state as a function of time (additional details in the
Materials and Methods) [37–38]. This procedure is similar to
characterizing the consensus behaviour of a population of infected
hosts that exhibit individual-to-individual variation.
To construct the single infection models, we formulated the
Boolean transfer functions from the current knowledge of the
immune regulatory processes and in case of ambiguity we
iteratively modified the transfer function by comparing the
simulated dynamic output with our empirical results on single
infection and with immune knockout studies (a detailed example is
reported in the Materials and Methods). Finally, to examine the
relative importance of the immune components, we perturbed
each node by setting their status to OFF and monitored parasite
activity up to the time-step required for parasite clearance/
reduction in the unperturbed system. Any increase in the infection
activity following the knockout of an immune node -which may
cascade to the connected downstream nodes- indicated the
importance of this node for parasite clearance. Nodes whose
deactivation led to long term persistence, represented by parasite
activity equal to 1, were classified as essential for clearance. This
procedure allowed us to mimic laboratory experiments of single
immune component knockouts and to follow the consequences on
parasite clearance.
B. bronchiseptica single infection
The onset of B. bronchiseptica infection in the lungs was simulated
by setting the state of the bacteria node ON and the state of the
nodes of the immune response OFF (Fig. 3A). As the infection
proceeded, and consistent with our empirical work [28], IFNc and
IL10 expression rapidly peaked and then slowly decreased below
the threshold through the course of the infection (Fig. 3B). B.
bronchiseptica has been suggested to induce IL10 production by T
cell subtypes, which inhibits IFNc in the lower respiratory tract
[25]. By explicitly including the bacteria mediated up-regulation of
IL10, through the type III secretion system (TTSS) modulation of
T
regulatory
cells
(Treg),
we
were
able
to
capture
the
establishment of the bacteria in the lungs followed by their
immune-mediated reduction and clearance. Activation of B cells
by T helper cells led to the prompt increase of peripheral
antibodies (serum IgG and IgA), in line with empirical data [26–
27,39]. Serum IgG reached and maintained long-lasting above-
threshold saturation in all simulations whereas IgA activity
dropped along with B. bronchiseptica and was turned off after 15
Figure 2. Network of immune components considered in single T. retortaeformis infection. Grey nodes have been quantified in the single
laboratory experiment. Abbreviations: IS: Larvae; AD: Adult; IL4II: Interleukin 4 in the systemic compartment; NE: Recruited neutrophils; IgA:
Antibody A; IL4I: Interleukin 4 in the small intestine; Th2II: Th2 cells in the systemic compartment; Th2I: Th2 cells in the small intestine; IgG:
Antibody G; IgE: Antibody E; IL10I: Interleukin 10 in the small intestine; IFNcI: Interferon gamma in the small intestine; IL12II: Interleukin 12 in the
systemic compartment; BC: B cells; DCII: Dendritic cells in the systemic compartment; DCI: Dendritic cells in the small intestine; Th1I: T helper cells
subtype I in the small intestine; PIC: Pro-inflammatory cytokines; Th1II: T helper cells subtype I in systemic compartment EC: Epithelial cells the small
intestine; T0: Naı¨ve T cells; EL2: recruited eosinophils; EL: resident eosinophils; IL13: Interleukin 13; IL5: Interleukin 5. Additional details in Figure 1.
doi:10.1371/journal.pcbi.1002345.g002
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
4
January 2012 | Volume 8 | Issue 1 | e1002345
time-steps (see Materials and Methods) (Fig. 3C). The rapid
recruitment of peripheral neutrophils to the lungs was possible
through pro-inflammatory cytokine mediated signalling (Fig. 3D),
while macrophages were recruited by IFNc secreted by Th1 cells.
The activation of neutrophils and macrophages by antibodies, via
the antibody-antigen complex and complement nodes (see Fig. 1),
led to bacterial phagocytosis and clearance from the lungs within
20 time steps, in agreement with our empirical work.
The relative importance of the different immune components
was then explored by knocking off single nodes and monitoring the
level of bacterial intensity at the 20th time-step, the time required
for B. bronchiseptica clearance from the lungs in the unperturbed
system. The perturbation results reproduced the observations from
B. bronchiseptica infections in the respective empirical knockout
experiments (Fig. 4A) [8]. For example, it has been observed that
B. bronchiseptica can persist in large numbers in mice where T0,
Th1 or B cells are depleted [8]; the key role of these nodes was
confirmed by our model. The simulations also highlighted the
crucial
role
of
pro-inflammatory
responses,
dendritic
cells,
macrophages and IL12 as their inactive state resulted in bacterial
persistence (Fig. 4A). In contrast, knocking out IL4 or any of the
15 remaining nodes of the network did not increase the activity of
the node Bordetella.
T. retortaeformis single infection
The infection of T. retortaeformis was simulated by setting the
state of the infective larvae node ON and the immune nodes OFF
(Fig. 5A). Ingested larvae were either killed by eosinophils, in a
Figure 3. Results of the simulations of the time course of the single B. bronchiseptica infection. Activity profiles (the probability of the
node being in an ON state at a given time-step) are reported for: A- Bacterial colonies in the lungs. B- Cytokines, IFNc, IL4 and IL10, in the lungs. C-
Serum antibodies. D- Peripheral neutrophils.
doi:10.1371/journal.pcbi.1002345.g003
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
5
January 2012 | Volume 8 | Issue 1 | e1002345
probabilistic manner [40–41], or successfully developed into
adults. Adults started to appear after 2 time-steps, mimicking the
natural development of infective third stage larvae into adults.
Following the infection, IFNc rapidly peaked after two time steps
while IL4 and IL10 activation followed with a delay, in line with
empirical findings (Fig. 5B) [31]. The initial vigorous expression
of IFNc was driven by dendritic cells, probably as an inflammatory
response to the infiltration of microflora and bacteria into the
damaged mucosa during the establishment of larvae [31]. This was
modelled by turning the activity of the local IFNc ON if
sufficiently stimulated by dendritic cells; the subsequent IFNc
activation occurred through a Th1 cell response. For the
interpretation of Fig. 5B, the fraction of IFNc activity that
occurred from 0 to 1 was due to a Th1 response while above 1 was
caused by dendritic cells. Dendritic cells also activated the Th2 cell
mediated expression of IL4 and as this arm of the immune
response developed, IFNc decreased although remained in an
active state throughout the infection (Fig. 5B). IL10 expression
was relatively low and similar to IL4, as found in our experimental
results. Naı¨ve T cell-initiated B cell proliferation stimulated the
prompt increase of mucus IgA, IgE and IgG above the activation
threshold (Fig. 5C). The consequent recruitment of neutrophils,
along with IgG, led to the reduction but not clearance of adult
helminths, consistent with the empirical observation that a few
individuals still harboured helminths in the duodenum at 120 days
post infection (Fig. 5A). Unlike IgA, whose activity followed the
dynamics of T. retortaeformis abundance, IgG activity remained
persistently high. In contrast to the small and short-lived neutrophil
peak, the eosinophil activity was higher and lasted longer (Fig. 5D).
The stability of the immune pathways and the reliability of our
parsimonious model were explored by systematically knocking out
network nodes and examining the effects on the activity of the
adult helminth node at the 20th time-step, the time point when the
unperturbed system reaches equilibrium (Fig. 4C). None of the
perturbations led to an activity of the adult parasite node of less
than 0.3, indicating that T. retortaeformis persists in the rabbit and
this is a robust outcome of the model, which matches our empirical
observations. Simulations suggested that the individual knockout
Figure 4. Parasite activity at the 20th time step from simulations where network nodes were individually knocked out (from 100
replicates). A- B. bronchiseptica in single infection. B- B. bronchiseptica in co-infection. C- T. retortaeformis in single infection. D- T. retortaeformis in
co-infection. Explanation of the abbreviations is reported in Figure 1, Figure 2 and Text S1.
doi:10.1371/journal.pcbi.1002345.g004
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
6
January 2012 | Volume 8 | Issue 1 | e1002345
of 14 nodes, including pro-inflammatory cytokines, IL13, naı¨ve T
cells, dendritic cells, eosinophils and neutrophils led to helminth
persistence in all the simulations (i.e. adult activity equal to 1)
(Fig. 4C). Interestingly, deletion of either local or systemic IL4
(IL4I or IL4II) reduced parasite activity, as IL4 contributed to
inhibit neutrophils (via the inhibition of the IL12 node). To
identify the nodes that may lead to faster reduction or clearance of
T. retortaeformis we constitutively turned ON single nodes. Over-
expression of recruited eosinophils, IL5, neutrophils and Th2 cells
in the small intestine reduced parasite activity below 0.5 (results
not shown). These and the knockout simulations suggested that
neutrophils and eosinophils are critically involved in the clearance
of T. retortaeformis infection.
B. bronchiseptica-T. retortaeformis co-infection
Network modelling.
To explicitly quantify the interactions
between B. bronchiseptica and T. retortaeformis the two single immune
networks were connected and the co-infection network simulated
as a single entity without changing the Boolean rules built for the
single networks, except for the adjustments necessary for assembly
(Fig. 6). The link between networks was established through the
cytokines, which maintain the communication between the
systemic and local immune processes as well as the cross-
interactions between infections. Specifically, we assumed a single
unlimited pool of naive T cells and three pools of cytokines: a pool
in the lungs, a pool in the small intestine (duodenum) and a
systemic pool interacting with both infections. For example, we
Figure 5. Results of the simulations of the time course of the single T. retortaeformis infection. Activity profiles (the probability of the node
being in an ON state at a given time-step) are reported for: A- Third stage infective larvae (L3) and adults. B- Cytokines, IFNc, IL4 and IL10 in the
duodenum. C- Mucus antibodies against helminth adult parasites. D- Peripheral eosinophils and neutrophils. Note that the IFNc concentration range
is between 0–2 to describe additional non-immune mediated activation of that node by the tissue damage (details in the Results).
doi:10.1371/journal.pcbi.1002345.g005
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
7
January 2012 | Volume 8 | Issue 1 | e1002345
assumed that only one pool of IL4 and IL12 exists in the systemic
compartment although antigen specific cells, polarized towards
bacteria or helminths, can produce these cytokines. In other words,
IL12 induced by bacterial factors can inhibit IL4 production by
helminth-specific Th2 cells. Local cytokine expression can be
affected by mucosal immune components, parasite intensity and the
systemic cytokine response. These assumptions allowed us to take
into account the compartmentalization of the infections (i.e. lungs
and duodenum) as well as bystander effects of the immune response
and the balance of the immune system as a whole. The dynamics of
the simulated immune components and associated parasite activity
were then compared with the empirical co-infection results.
B.
bronchiseptica.
Simulations
showed
the
switch
of
cytokines from the initial high expression of IFNc and IL10 to
the late increase and long activity of IL4 (Fig. 7B). Antibodies
quickly increased, serum IgG remained consistently high while IgA
decreased below the threshold after 5 time-steps as bacterial
numbers declined (Fig. 7C). The peripheral neutrophil activity
was higher in co-infected compared to single infected hosts,
however, their recruitment in the lungs was completely turned off
after 14 time steps (Fig. 7D vs Fig. 5D). These temporal patterns
resulted from the inflammatory cytokines produced in response to
both T. retortaeformis and B. bronchiseptica and should be interpreted
as a mixed activity against both parasites. Our simulations
indicated similarities between B. bronchiseptica single and co-
infection, such as the rapid increase in systemic IgA, IgG and
neutrophils but also differences, namely, the higher and longer
activity of IL4 in the lungs and the longer presence of peripheral
neutrophils in dual compared to single infection. Overall, despite a
few immunological differences the dynamics and timing of B.
bronchiseptica clearance in the lungs of co-infected hosts was similar
to that observed in the single infection and driven by phagocytic
cells activated by antibodies and Th1 cells (Fig. 7A). The low but
non-zero activity of bacteria in the co-infection steady state
indicated that the infection was not cleared in a small fraction of
the replicate simulations (8%) (Fig. 7A). Specifically, IL4 activated
by eosinophils in response to T. retortaeformis was responsible for the
persistence of bacteria in the lungs. During single bacterial
infection the IL4 level was relatively low and controlled by the
inhibitory effect of IL12, however, during the co-infection this
suppressive effect was not observed as a Th2 environment
dominated. This model prediction is supported by previous
studies that showed a delayed bacterial clearance in case of
persistent IL4 [42]. Knockout perturbation analysis confirmed that
IL4 produced by eosinophils was responsible for this occasional
bacterial persistence, since the deletion of this node led to the
complete clearance of the infection in all the simulations (Fig. 4B).
Bacterial persistence was also observed when Th1 cells, antibodies,
pro-inflammatory cytokines or the activated phagocytes node were
individually knocked out. The 15 nodes whose deletion had very
little effect in the single infection had a similarly weak effect on
bacterial numbers in the co-infection (Fig. 4A vs 4B). Interestingly
and contrary to the single infection, the knockout of bacteria-
activated epithelial cells did not influence B. bronchiseptica activity
since the pro-inflammatory cytokines node, which is downstream
of the epithelial cells node, was also activated by the helminths.
This between-organ communication was possible by assuming a
single pool of cytokines and their free movement among organs,
for example via the blood system. Perturbation of any of the 17
helminth-specific nodes had a generally weak effect on bacterial
activity.
T. retortaeformis.
The concurrent effect of B. bronchiseptica
on T. retortaeformis infection dynamics was equally examined.
Counter to our initial predictions, lower establishment and faster
clearance of T. retortaeformis were observed in co-infected compared
to single infected hosts (Fig. 8A vs 5A). The model showed high
activities of IFNc and IL10 and low expression of IL4 (Fig. 8B).
As observed in the single infection, the early peak of IFNc (having
activity .1) was caused by an initial host-mediated inflammatory
response, as an immediate-type hypersensitivity reaction of the
tissue to the establishment of infective larvae. This local activation
was then followed by a Th1 mediated IFNc expression, consistent
Figure 6. Network of immune components considered in the B. bronchiseptica-T. retortaeformis co-infection. Bi-directional black arrows
indicate the influence of components from one network on the common cytokine pool and vice a versa.
doi:10.1371/journal.pcbi.1002345.g006
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
8
January 2012 | Volume 8 | Issue 1 | e1002345
with the single infection model. A bystander Th1 mediated effect
of B. bronchiseptica synergistically contributed to this pattern by
enhancing the activity and duration of IFNc expression in the
duodenum. Simulations suggested that the local IL10 expression,
higher in the dual compared to the single infection, was a
bystander effect induced by the type three secretion system (TTSS)
of B. bronchiseptica through Treg cells. Also, the early IL4 expression
was suppressed by the Th1 mediated IFNc phenotype activated
both by the helminth, during the initial establishment, and the
bacterial co-infection. Mucus IgG remained consistently active
from time step 3 while mucus IgA was at the highest between 5
and 10 time steps but decreased thereafter (Fig. 8C). Recruited
peripheral neutrophils but not eosinophils were higher in the dual
infection compared to single helminth infection simulations
(Fig. 8D).
To provide a parsimonious mechanism that could explain the
rapid helminth clearance, the immune nodes of the co-infection
network were systematically knocked out and the helminth activity
examined at the 20th time-step (Fig. 4D). Similar to the single
infection, the deactivation of key nodes, for instance B cells,
dendritic cells or T cells, resulted in helminth persistence in all the
simulations (adult activity equal to 1). Unlike in the single
infection, knockout of resident eosinophils or the IL12II node
did not lead to helminth persistence. This was because the
induction of downstream processes, such as the activation of IL4
or IFNc was now performed through the complementary effect of
the bacterial nodes and their bystander effects. Interestingly, the
single knockout of 92% of the nodes, including bacterium-specific
nodes, increased helminth activity, compared to the unperturbed
co-infection model, but did not lead to helminth persistence in
Figure 7. Results of the simulations of the time course of B. bronchiseptica from the co-infection. Activity profiles (the probability of the
node being in an ON state at a given time-step) are reported for: A- Bacterial colonies in the lungs. B- Cytokines, IFNc, IL4 and IL10, in the lungs. C-
Serum antibodies. D- Peripheral neutrophils.
doi:10.1371/journal.pcbi.1002345.g007
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
9
January 2012 | Volume 8 | Issue 1 | e1002345
every simulation. From a modelling perspective, the network in
Fig. 6 represents a sparse causal model of co-infection dynamics.
In other words, all these nodes or nodes downstream of the
targeted nodes contribute to, but are not required for, T.
retortaeformis clearance. The knockout of effector nodes namely,
recruited eosinophils or neutrophils and cytokines like IL5 or IL13,
resulted in helminth long term persistence, supporting the
hypothesis that a co-operative mechanism including leukocytes,
antigen-specific antibodies (IgG and IgE) and Th2 mediated IL5
and IL13 are critical in helminth clearance [43–49]. The role of
IL5 and IL13 is mostly in the recruitment of eosinophils while
neutrophils are recruited by pro-inflammatory cytokines and Th1
mediated IFNc. Though antibodies recognize the helminth, in this
model they do not form complexes, rather, they attract leukocytes
bearing Fc-receptors leading to the recruitment of neutrophils and
eosinophils.
A comparison between single and dual infection offers insights
into the contribution and balance of these two leukocytes to T.
retortaeformis dynamics. In the single infection, when neutrophils are
only transiently activated, the recruited eosinophils were relatively
more important to parasite reduction, although they were not
sufficient to clear the infection. In the co-infection, the robust and
early
activation
of
recruited
neutrophils
-which
decreased
following helminth reduction- and the activation of recruited
Figure 8. Results of the simulations of the time course of T. retortaeformis infection from the co-infection. Activity profiles (the
probability of the node being in an ON state at a given time-step) are reported for: A- Third stage infective larvae (L3) and adults. B- Cytokines, IFNc,
IL4 and IL10 in the duodenum. C- Mucus antibodies against adult helminths. D- Peripheral eosinophils and neutrophils. Note that the IFNc
concentration range is between 0–2 to describe additional non-immune mediated activation of that node by the tissue damage (details in the
Results).
doi:10.1371/journal.pcbi.1002345.g008
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
10
January 2012 | Volume 8 | Issue 1 | e1002345
eosinophils -which are important in reducing the number of
infecting larvae and are required for neutrophils to successfully
reduce the helminths- highlighted the synergistic role of these cells
in the observed fast clearance of T. retortaeformis. To explicitly study
the bacterial components inducing these two leukocytes, we
switched the nodes to ON one at a time and found that dendritic
cells and Th1 cells, activated by bacteria, led to a significant
increase in neutrophil activity (results not shown). Counter to this,
no bacterial nodes significantly contributed to eosinophil produc-
tion. Switching ON the type III secretion system node transiently
increased eosinophil activity, compared to the unperturbed
system, as expected from the role of TTSS in the induction of
Th2 related cytokines [25]. However, this had a very short lived
effect since TTSS was neutralized by antibodies. In summary,
simulations suggest that strong inflammatory responses generated
by the bacteria led to an early increase of neutrophils which
contributed to a prompt and more effective helminth reduction.
Empirical co-infection experiment
A B. bronchiseptica-T. retortaeformis co-infection experiment was
carried out and the empirical results were used to validate the co-
infection dynamic model. A statistical analysis was also performed
between the single and co-infection trials to further reinforce our
modelling outputs. However, while the statistical findings provide
an insight into the relationships among the immune components,
no mechanistic understanding or dynamic outcomes can be
established between these variables and parasite abundance. The
network-based discrete dynamic models allowed us to establish
such connections and causal interactions between the various
components. Overall, we found that the parsimonious dynamic
model correctly predicted the observed dynamics of concurrent B.
bronchiseptica and T. retortaeformis co-infection.
B.
bronchiseptica.
The
bacterial
colonization
of
the
respiratory tract of co-infected rabbits was similar to single
infection. B. bronchiseptica abundance in the lungs increased in the
first 7 days post challenge and decreased thereafter, as seen in the
dynamic model; by 90 days bacteria were completely cleared from
the lungs and trachea but persisted in the nasal cavity (Fig. 9A).
Based on the a priori measurement of optical density with a
spectrophotometer, individuals received a dose similar to the single
infection however, the a posteriori quantification of bacteria on
blood agar plates suggested that an inoculum of 10,600 CFU/ml
was administered, five times less than the single infection dose
[28]. If we consider the second measure correct, the lower dose did
not affect replication and the colony numbers quickly reached
values comparable to single infection by 3–7 days post challenge.
Specifically, the average number of bacteria in the lower res-
piratory tract was analogous to the single infection but significantly
higher numbers were observed in the nasal cavity during the
infection (Fig. 9A, Table 1). Confirming the model simulations,
IFNc quickly increased, peaked by 3 days post challenge and
quickly decreased thereafter. IL10 followed a similar pattern with
a small delay while IL4 slowly increased and peaked 60 days post
infection (Fig. 9B). Serum antibodies showed a trend similar to
that of the single infection, in accordance with our dynamic model.
IgG
rapidly increased
and remained
high throughout
the
experiment while IgA rapidly decreased although a second peak
was observed around week twelve, this second peak was based on
much fewer individuals and, probably, it was not biologically
relevant (Fig. 9C). Peripheral leukocytes concentration reflected
the response to both infections specifically, neutrophil numbers
showed a robust peak at week three while eosinophil numbers
increased between two and five weeks post-infection, both in
agreement with the model (Fig. 9D and Fig. 10D).
A combination of principal component analysis (PCA) and
generalized linear models (GLM) indicated that B. bronchiseptica in
the lungs was negatively associated with IL4, serum IgG and IgA
(PCA axis 1), and peripheral eosinophils and neutrophils (PCA 2,
Table S1). To compare the immune response between single and
co-infected hosts, variables were scaled over the controls. Co-
infected rabbits exhibited higher IL4 (coeff6S.E. = 20.87960.210,
P,0.001), serum IgG (0.16660.043 P,0.001) and neutrophils
(0.23360.050, P,0.0001) but lower eosinophils (21.70560.006,
P,0.0001) compared to single infected individuals. It is important
to note that a low or negative cytokine Ct value (cycle threshold
scaled over the controls) identifies high mRNA expression and vice
versa, thus in the models low Ct values are translated as high
cytokine activity. The remaining variables were not significant,
although this should not be interpreted as a complete lack of
variability between the two infections. Indeed, as highlighted in the
network model these variables play a secondary but still necessary
role in generating immune differences between infections.
T. retortaeformis.
Helminth intensity significantly decreased
with the progression of the infection and organ location (high
numbers in the duodenum, SI1, and low in the ileum, SI4)
however, counter to our expectation and consistent with our
model simulations, lower establishment and faster clearance were
observed
in
co-infected
compared
to
single
infected
hosts
(Fig. 10A, Table 2). As predicted by our dynamic model,
strong and persistent IFNc expression but relatively low IL4 and
IL10 were found in the duodenum of infected rabbits compared to
the controls (Fig. 10B). Consistent with the single infection and
the dynamic model, mucus antibody quickly increased, IgG
remained relatively high for the duration of the trial while IgA
declined from day 30 post challenge (Fig. 10C). The peripheral
leukocyte profile has already been described in the bacteria section
(Fig. 9D and Fig. 10D). Principal component analysis identified
that T. retortaeformis was positively associated with the first axis
(PCA 1), mainly described by the interaction among the three
cytokines, and negatively related to the second axis (PCA 2)
represented
by
eosinophils
and
antibodies
(Table
S2).
Interestingly, cytokines were positively correlated (IFNc vs IL10:
r = 58% P,0.001; IL4 vs IL10: r = 54%, P,0.01), indicating the
co-occurrence of a specific response to the helminth, through IL4,
but
also
a
robust
inflammatory/anti-inflammatory
reaction
probably
caused
by
the
parasite
damaging
the
mucosal
epithelium and resulting in bacterial tissue infiltration during
larval establishment [31]. The comparison of immune variables
between single and co-infection showed higher neutrophils
(P,0.0001) and a tendency for higher IL10 (P = 0.058) in co-
infected compared to single infected hosts. The overall expression
of IL4 was lower in co-infected individuals (P = 0.035), however
higher values were observed at 14 days post infection (interaction
of IL4 with day 14 post infection P = 0.046).
Discussion
Co-infections affect the immune responses but how the systemic
processes interact and influence the kinetics at the local sites of
infection is
still
unclear.
The
majority
of studies
on the
immunology of co-infection have focused on either one of the
infecting species or a restricted class of cells or immune processes,
and often concentrated on the early stage of the infection [4,9–
14,50–52]. These studies have been extremely useful in highlight-
ing not only the similarities across systems but also the specificity of
some of these mechanisms and how they differ from single
infections. Yet, there is a need for a comprehensive understanding
of these processes as a whole individual response, how systemic
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
11
January 2012 | Volume 8 | Issue 1 | e1002345
and localized processes interact and how they dynamically evolve
during the course of the co-infection. We used a combination of
laboratory experiments and modelling to examine the dynamic
network of immune responses to the respiratory bacterium B.
bronchiseptica and the gastrointestinal helminth T. retortaeformis. Our
aim was to identify the parsimonious processes and key cells
driving parasite reduction or clearance and how they changed
between single and co-infections.
We confirmed the initial hypothesis of immune mediated
interactions between the two parasites, however, our initial
predictions were only partially supported. The most unexpected
result was the faster clearance of T. retortaeformis in co-infected
compared to single infected individuals, which was observed in the
model simulations and confirmed in the empirical data. Neither
did we expect to find that B. bronchiseptica infection in the lungs was
not significantly altered by the concurrent helminth infection,
despite the increase in local IL4 expression observed in both the
simulations and the experiment. We found a small difference in
bacterial clearance between single and co-infection (Fig. 3A vs
Fig. 7A) and we were able to explain that this was driven by the
differential recruitment of phagocytes, particularly macrophages
induced by IFNc during co-infections, as compared to the single
infection. However we found that T. retortaeformis enhanced
individual variability in the immune response to B. bronchiseptica
infection by occasionally reducing the overall efficacy of the Th1
immune
response,
through
eosinophil
produced
IL4,
and
Figure 9. Summary of B. bronchiseptica intensity and immune variables from the experimental co-infection. Mean6SE during the course
of the infection (days or weeks post infection) are reported. A- Bacterial intensity in the respiratory tract. For comparison, empty black circles
represent the bacterial intensity in the lungs from the single infection. B- Cytokines, IFNc, IL4 and IL10 in the lungs. C- Anti-bacterial IgA and IgG in
serum. D- Peripheral neutrophils. For C and D, infected hosts: full circles, controls: empty circles.
doi:10.1371/journal.pcbi.1002345.g009
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
12
January 2012 | Volume 8 | Issue 1 | e1002345
preventing bacterial clearance in the lungs, a pattern observed in
8% of the simulations. The helminth mediated delay or absence of
bacterial clearance from the lower respiratory tract was indeed our
original hypothesis and interestingly the model indicated that this
is still a possible outcome of the interaction between these
parasites. This implies that heterogeneities in the host immune
response are not exceptional events and can have major effects on
the dynamics of infection and persistence. Our model was able to
capture this variability because of the large number of simulations;
in other words a large group of infected individuals were examined
compared to our much smaller sample tested in the laboratory.
Follow-up experiments using a much larger number of animals or
replication of the same experiment a few times may lead to the
experimental observation of this behaviour. The empirical findings
also showed that T. retortaeformis infection resulted in a significant
increase of bacterial numbers in the nasal cavity compared to
single infection, particularly after the initial phase of the infection.
At the host population level these findings support the hypothesis
that co-infections can increase individual variability to infections
by altering bacterial intensity and prevalence, and this can have
major consequences for the risk of transmission and disease
outbreak [53]. Overall, our dynamic models indicated that the
clearance of B. bronchiseptica in single and co-infection was mainly
driven by phagocytosis of bacteria by macrophages and neutro-
phils activated by antibodies. Deactivating nodes that affected
bacterial recognition (e.g. pro-inflammatory cytokines, epithelial
cells or antibodies) or phagocytosis (e.g. Ag-Ab complex or
macrophages) increased bacterial abundance in single and dual
infections, suggesting that these cells are necessary for controlling
B. bronchiseptica.
The immune network for T. retortaeformis was less detailed than
that for the bacterial network, nevertheless, the model predictions
of the activity pattern of the helminth and the immune variables
that have been quantified were in agreement with our empirical
studies. To our surprise the prediction of no effect of B.
bronchiseptica on T. retortaeformis infection was proven wrong.
Simulations suggested that the combined effect of neutrophils,
eosinophils and antibodies (IgG and IgE) led to helminth
expulsion. Neutrophils and eosinophils were activated through
antigen-specific Th1 and Th2 responses, respectively. Th2-
mediated differentiation of progenitor eosinophils (i.e. resident
eosinophils), modulated by IL5 and IL13, also played an
important role in helminth reduction in single infection, as
indicated by the perturbation results. Previous studies on murine
systems have shown that IL13 can complement IL4 or play an
alternative or even stronger role in helminth infections [42–43].
Using our modelling approach we showed that IL5 and IL13 had
complementary abilities against helminths and contributed to
parasite reduction both in single and co-infection. The strategic
role of neutrophils in bacteria-helminth co-infections has been
previously described [44]; using a modelling approach not only we
confirmed
this
property
but
also
suggested
a
non-specific
infiltration of effector cells into infected tissues.
The mixed Th1/Th2 response in the duodenum was driven by
different processes. The early IFNc inflammatory signal observed
both in single and co-infection was a host response to the mucosa
damage by helminth establishment, and probably bacteria and
microflora infiltration from the lumen [31]. This was also
complemented by a bystander effect of B. bronchiseptica co-infection,
rather than a helminth induced up-regulation of this cytokine to
facilitate tissue colonization [31]. This mechanism is supported by
our recent studies on cytokine expression in different organs of
single and co-infected rabbits at seven days post infection, where
we showed that IFNc was remarkably reduced in the ileum,
mesenteric lymph node and spleen, where fewer or no helminths
were found, compared to the duodenum [54]. The Th2 cell
activity was primarily focused on preventing parasite establishment
and survival. These findings indicate that these two cytokines are
not mutually exclusive but can simultaneously act on different
tasks specifically, tissue repair, inflammatory response to micro-
flora
infiltration
and
helminth
clearance.
Mixed
Th1/Th2
phenotypes are not new to parasite infections and the murine-
Schistosoma mansoni or Trichuris muris systems are well described
examples [55–57].
Model strengths and limitations
The aim of this study was to develop tractable dynamic models
that could capture the interactions of multi-organ, multi-species
co-infection immune processes as well as single infection dynamics.
We found the discrete dynamic Boolean models a feasible and
reliable approach for this task since we lacked accurate spatio-
temporal details on the majority of the variables and the kinetic
parameters required to develop robust quantitative, differential
equation-based models [34–36]. Boolean models assume that what
matters the most is whether the concentration or level of
expression of a node (i.e. immune cell) is higher or lower than
an a priori fixed threshold. They also use a parameter-free
combinatorial description for the change in status of the nodes,
thus avoid the need for parameter estimation while being
sufficiently flexible. Indeed, Boolean models have been successfully
used in a variety of contexts, from signal transduction [38,58] to
development [59–60], immune responses [8,61–62] and popula-
tion-level networks [63]. Choosing a quantitative modelling
approach would have forced us to drastically simplify our system,
impose a large number of assumptions on the concentration,
transfer function and kinetic parameter of each node, and so we
would have not been able to offer robust predictions on the role of
many immune components and on how they affect the dynamics
of parasite infection in our system.
Table 1. Summary of linear mixed effect model (LME)
between B. bronchiseptica abundance (CFU/g), as a response,
and infection type (single or co-infection), day post infection
(DPI) and organ (lung, trachea or nose) as independent
variables.
Coeff±S.E., d.f.
P
Intercept
14.48360.745, 122
0.00001
Infection type
1.71160.895, 60
0.061
Trachea
20.25960.661, 122
0.695
Nose
0.72160.757, 122
0.343
DPI
20.11360.010, 60
0.00001
Infection type*DPI
20.04560.015, 60
0.005
Trachea*DPI
20.00860.010, 122
0.425
Nose*DPI
0.07660.011, 122
0.00001
Infection type*Trachea*DPI
0.00460.013, 122
0.745
Infection type*Nose*DPI
0.03960.015, 122
0.009
AIC
1022.895
Host ID random effect (intercept S.D.)
1.113
AR(1)
0.311
The random effect of the host identity code (ID) and the autocorrelation effect
(AR-1) of sampling different organs for the same host are also reported.
doi:10.1371/journal.pcbi.1002345.t001
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
13
January 2012 | Volume 8 | Issue 1 | e1002345
Our models were based on the most updated knowledge of the
immune components and processes during single infections to
Bordetella and gastrointestinal helminths. In cases of uncertainty
(e.g. whether two co-regulators were independent or synergistic)
we tested a number of different assumptions (i.e. Boolean transfer
functions) and selected the function that best described our single
infection experiments in terms of the: timing of events, node
activities and importantly, parasite steady state (see Materials and
Methods for an example). To overcome the fact that timescales
and duration of immune processes were unknown, we generated
repeated simulations with various update orders, which essentially
allowed us the sampling of various time durations and probing
which model output was robust to timing uncertainties. Impor-
tantly, the outputs of our simulations were not averages but the
quantification of the agreement between runs, for example, the
anti-B. bronchiseptica IgG activity of 1 after step 4 in Fig. 3C means
that following this time point all runs show an above-threshold
concentration of IgG regardless of timing variations. By compar-
ing the features of the curves (e.g. saturating shape, peak
occurrence and timing) with our experimental observations we
were able to confirm the accuracy of the model in predicting the
observed kinetics.
Figure 10. Summary of T. retortaeformis intensity and immune variables from the experimental co-infection. Mean6SE during the
course of the infection (days or weeks post infection) are reported. A- Helminth intensity in the small intestine sections, from the duodenum (SI-1) to
the ileum (SI-4), respectively. The helminth development during the course of the infection is as follows: 4 days post infection (DPI) third stage
infective larvae (L3), 7 DPI both L3 and fourth stage larvae (L4), from 14 DPI onwards adult stage only. For comparison, empty black circles represent
the helminth intensity in the duodenum from the single infection. B- Expression of cytokines, IFNc, IL4 and IL10 in the duodenum. C- Mucus antibody
against adult helminths, IgA (C1) and IgG (C2), from the duodenum to the ileum. D- Peripheral eosinophils. For C and D, infected hosts: full circles,
controls: empty circles.
doi:10.1371/journal.pcbi.1002345.g010
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
14
January 2012 | Volume 8 | Issue 1 | e1002345
One of the strengths of our modelling was to make predictions
on the dynamics of parasite clearance based on the perturbation of
the nodes (i.e. single node knockout). These simulations followed
the classical knockout lab experiments where single immune
components (nodes) were turned off from the beginning of the
simulation and the dynamics of the immune response, as well as
parasite clearance, were examined. This approach allowed us to
explore the knockout of a large number of immune variables,
determine the most important components modulating the
immune response and highlight how they differed between single
and co-infection. These findings can be tested in the laboratory by
performing knockout experiments of the crucial immune variables
in different infection settings. For example, we can block
neutrophil production or the cytokine IL13 and examine whether
helminths persist -as predicted by our knockout simulations- or are
slowly cleared in bacteria co-infected rabbits. Similarly, we can test
the predicted different response of knocking out IL4 in helminth
and bacteria-helminth co-infection, specifically, whether clearance
is higher than in un-manipulated individuals in single helminth
infection and lower than in un-manipulated co-infected hosts. We
should also pay more attention to B. bronchiseptica infection in the
nasal cavity and develop dynamic immune models that can
explain bacterial persistence as well as possible clearance under
different knockout scenarios both in single and co-infection. The
most
parsimonious
hypotheses
can
then
be
tested
in
the
laboratory. This is important because our recent work suggested
that bacterial shedding during the long lasting chronic phase relies
mainly on the infection of the upper respiratory tract, once it has
been cleared from the lungs and trachea [28]. This has relevant
epidemiological implications for bacterial transmission that go
beyond the rabbit-parasite system. We can further refine our
models and explore the dynamics of the parasite-immune network
when the onset of the co-infections is lagged between the parasite
species or one parasite is trickle dosed, a dynamic that resembles
more closely to the natural conditions. Again, these predictions
can be validated through experimental infections of naı¨ve or
knockout animals. It is important to underline that our approach
can be adapted to a large variety of bacteria-helminth co-
infections of many host systems where organ compartmentaliza-
tion, differences in the time of infection or number of parasite
stages are observed.
In conclusion, we showed that network-based discrete dynamic
models are a useful approach to describe the immune mediated
dynamics of co-infections. These models are robust as well as
sufficiently tractable to qualitatively capture the complexity of the
immune system and its kinetics over time. Arguably, the main
limitation of our modelling approach is that it lacks a fully
quantitative component. Yet, this work demonstrated that it is
possible to build comprehensive qualitative dynamic models of the
local and systemic immune network of single and co-infection that
are validated by empirical observations. Importantly, this study is a
fundamental starting point towards the future construction of
quantitative models based on simplified networks that describe the
kinetics and intensities of the causal relationships among key
immune
components
identified
in
qualitative
models.
Our
approach showed that we can refine the conventional approach
of using the Th1/Th2 paradigm, by identifying system-specific
functions or cell groups that can capture crucial immune processes
during co-infections. While our parsimonious dynamical models
were able to capture the patterns of single and co-infection
observed in the experiments, we are aware that they are far from
complete in describing the immunological complexity of the
processes involved and cells activated. Nevertheless, they provide a
parsimonious description of the system that can be experimentally
tested. Ultimately, we showed that we cannot predict how the
immune system reacts to co-infections based on our knowledge of
single infection. More needs to be done to clarify the immune
mechanisms involved in bacteria-helminth co-infections and how
individual hosts balance the immune system as a whole.
Materials and Methods
Network modelling
Network assembly.
Interaction networks were built from
the available literature and adapted to our system. Bacteria,
helminth and the components of the immune system (i.e. immune
cells
and
cytokines)
were
represented
as
network
nodes;
interactions, regulatory relationships and transformations among
components were described as directed edges starting from the
source node (regulator) and ending in the target node. We
incorporated regulatory relationships that modulate a process (or
an unspecified process mediator) as edges directed toward another
edge. The regulatory effect of each edge was classified into
activation or inhibition, visually represented by an incoming black
arrow or an incoming red blunt segment. Since not all processes
involved in natural B. bronchiseptica and T. retortaeformis infections
are known or generally addressed in the rabbit infection model, we
extended
the
set
of
known
interactions
following
general
immunological knowledge on bacterial and helminth infections.
We constructed three networks: two networks that describe the
respective single infections and one that links the first two and
represents a co-infection network. A detailed description of each
network is given below.
B. bronchiseptica single infection.
Infection of the lungs
starts with the node Bacteria that leads to a cascade of immune
Table 2. Summary of linear mixed effect model (LME)
between T. retortaeformis abundance (worm/small intestine
length) as a response, and infection type (single or co-
infection), day post infection (DPI) and organ location (from
the duodenum -SI1- to the ileum -SI4-), as independent
variables.
Coeff±S.E., d.f.
P
Intercept
3.04460.178, 207
0.00001
Infection type
20.77860.240, 68
0.002
SI-2
20.52660.103, 207
0.001
SI-3
21.76660.138, 207
0.00001
SI-4
22.53060.159, 207
0.00001
DPI
20.02960.003, 68
0.00001
Infection type*SI-2
0.04560.109, 207
0.682
Infection type*SI-3
0.24360.146, 207
0.097
Infection type*SI-4
0.52060.169, 207
0.001
Infection type*DPI
0.00560.004, 68
0.214
SI-2*DPI
0.00360.001, 207
0.023
SI-3*DPI
0.01660.002, 207
0.00001
SI-4*DPI
0.02360.002, 207
0.00001
AIC
500.453
Host ID random effect (intercept S.D.)
0.001
AR(1)
0.773
The random effect of the host identity code (ID) and the autocorrelation effect
(AR-1) of sampling different organs of the same host are also reported.
doi:10.1371/journal.pcbi.1002345.t002
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
15
January 2012 | Volume 8 | Issue 1 | e1002345
interactions (Fig. 1, Text S1). This node includes generic
virulence factors of the bacteria such as the lipopolysaccharide
chain (LPS) required for tissue adherence following recognition of
bacteria by epithelial cells. Other bacterial virulence factors,
particularly O-antigen and type III secretion system (TTSS), are
explicitly included as separate nodes in the network and are
involved in the initial immune recognition of the bacteria node.
Upon detection, epithelial cells activate pro-inflammatory cyto-
kines, which in turn activate dendritic cells, often the most
important antigen presenting cells. Dendritic cells are also
activated by IFNc. Dendritic cells induce differentiation of naı¨ve
T cells (T0) by producing IL4 and IL12. The cytokine profile
along with the antigen leads to the activation of T cell subtypes
including helper and regulatory T cells. T helper cells are activated
in the lymph nodes (Compartment II) and subsequently tran-
sported to the site of infection (Compartment I). IL4 is also
produced by differentiated Th2 cells; IL4 and IL12 inhibit each
other and IL4 also inhibits IFNc. T regulatory (Treg) cells are
stimulated by the type III secretion system of B. bronchiseptica to
produce IL10. Th1 cells produce IFNc which along with pro-
inflammatory cytokines activates neutrophils and macrophages. A
different subtype of T cells, follicular T helper cells, is known to
stimulate B cell activation. To simplify the network we assumed
that naı¨ve T cells could play this role. Antigen-specific B cell
proliferation leads to the production of antibodies, namely IgG
and IgA. IgA production occurs only in the direct presence of
antigen unlike IgG that persists after bacterial clearance [28]. IgG
and bacteria complexes also induce complement fixation along
with bacteria themselves. Activation of complement by bacteria is
inhibited by O-antigen. The node ‘‘activated phagocytic cells’’
represents the outcome of the stimulation of neutrophils and
macrophages by antibody-antigen complex and complement.
These cells induce the node phagocytosis that depletes bacteria.
T. retortaeformis single infection.
The network starts with
infective larvae that develop into adults with no delay in the larval-
adult development, adults appear 2 time steps post infection
(Fig. 2, Text S1). Both parasite stages activate epithelial cells that
lead to the production of pro-inflammatory cytokines which then
activate dendritic cells and neutrophils, with the latter able to
inhibit adult helminths. Infective larvae stimulate IL13 production
by resident eosinophils and these recruit additional eosinophils
from the progenitor cells in the peripheral blood [63]. Eosinophils
can kill larvae through a stochastic process described by a uniform
distribution [64]. IL5 secreted by Th2 cells is required for the
recruitment of additional eosinophils. Infective larvae also directly
activate IFNc by damaging the mucosa tissue and causing a host
inflammatory response. This process does not include Th1 cells.
Pro-inflammatory cytokines activate dendritic cells that stimulate
naı¨ve T cells (T0). As described for B. bronchiseptica, dendritic cells
interact with naı¨ve T cells (T0) leading to the activation of T cell
subtypes Th1 and Th2 through the production of IL12 and IL4.
IL4 is also produced by Th2 cells and IL4 and IL12 inhibit each
other. Consistent with the bacteria network, the activation of T
helper cells occurs in the lymph nodes (Compartment II) and
subsequently transported to the site of infection (Compartment I).
In compartment I, IFNc is produced by Th1 cells and dendritic
cells.
IL4
and
IL10,
produced
by
Th2
cells,
have
anti-
inflammatory properties and inhibit pro-inflammatory cytokines
and neutrophils. Naive T cells stimulate clonal expansion of B cells
and these lead to the production of antibodies such as IgG. While
B cells can secrete IgG much longer after antigen removal, IgA
production is assumed to be in response to larval establishment
and development. The IgE isotype is produced upon signalling
from either IL4 or IL13. Among these antibodies IgG inhibits adult
helminths while IgE and IgA are involved in activating eosinophils
and inhibiting pro-inflammatory cytokines respectively.
B. bronchiseptica-T. retortaeformis co-infection.
The co-
infection immune network was developed by combining the two
single infection networks together (Fig. 6, Text S1). This network
is characterized by three compartments, representing the lungs,
the small intestine (duodenum) and the systemic compartment (e.g.
the lymphatic system). The connection of the networks and the
immune mediated interactions between parasites were represented
through the cytokines produced as a single pool. Local cells
activated by bacteria and helminths can contribute to cytokine
production, which are then transported through the blood and
disseminate to other organs [63]. For example, pro-inflammatory
cytokines are systematically detectable when any one of the
parasites activates epithelial cells. Similarly, IL4 or IL12 can be
produced by B. bronchiseptica- or T. retortaeformis-specific T subtypes
or dendritic cells. For the co-infection network, Tregs are induced
by bacteria which produce IL10 that can ultimately affect the
helminth, since IL10 is not an antigen-specific node. Moreover,
there is only a single pool of naı¨ve T cells that induces T cell
subtypes against either the bacteria or the helminths, depending
on the antigen-specific dendritic cells.
Discrete dynamic model implementation.
The immune-
parasite
interaction
networks
were
developed
into
discrete
dynamical models by characterizing each node with a variable
that can take the ON state, when the concentration or activity is
above the threshold level necessary to activate downstream
immune processes, or the OFF state when activity is below this
threshold. The evolution of the state of each node was described
by a Boolean transfer function (Text S1) [32]. Target nodes with a
single activator and no inhibitors follow the state of the activator
with a delay. The operator AND was used to describe a synergistic
or conditional interaction between two or more nodes that is
necessary to activate the target node. When either of the nodes
were sufficient for the activation of the target node we used the
operator OR. An inhibitory effect was represented by an AND
NOT operator. In cases where prior biological information did not
completely determine the transfer functions (e.g. there was no
information
whether
two
coincident
regulatory
effects
are
independent
or
synergistic),
different
alternative
transfer
functions were tested. The transfer functions that reproduced the
qualitative features of the single infection experimental time
courses, such as the parasite clearance profile, the relative peaks of
different cytokines or the saturating behaviour of IgG as compared
to IgA, were selected. For example, IL4 is produced by T helper
cells during T helper cell differentiation as well as by eosinophils in
response to stimulation by nematode antigens or allergens. While
IL12 is known to inhibit the production of IL4, there are two
possible ways this cytokine may interact with IL4: IL12 can inhibit
IL4 produced by T helper cells or IL12 can suppress IL4
production by blocking both the T helper and eosinophil signal.
The inhibitory effect of IL4 on the activation of neutrophils is
known. The two transfer functions were then examined by
comparing the temporal pattern for neutrophils and IL4 from the
single T. retortaeformis infection model with the experimental
observations. The second transfer function did not reproduce
the observed low activity of IL4 -compared to the other cytokines-
in the duodenum at day 14 post infection and it also led to higher
neutrophil activity, compared to the other leukocytes, than the
empirical data. Since the first transfer function did not lead to such
deficiencies, we chose the first over the second rule. The transfer
functions used in the co-infection model were the same as, or the
relevant composites of, transfer functions used in each individual
infection. Thus, the Boolean transfer functions applied in our
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
16
January 2012 | Volume 8 | Issue 1 | e1002345
model provide a mechanistic understanding of the interactions
leading to bacterial or helminth clearance.
The status of the system across time was simulated by repeatedly
applying the Boolean rules for each node until a stationary state
(e.g. clearance of the parasite) was found. Since the kinetics and
timescales of the individual processes represented as edges are not
known, a random order asynchronous update was selected
wherein the timescales of each regulatory process were randomly
chosen in such a way that the node states were updated in a
randomly
selected
order
during
each
time-step
[32].
The
asynchronous algorithm was: X t
i ~Fi(X ta
a ,X tb
b ,X tc
c ,:::), where F is
the Boolean transfer function, ta, tb, tc represent the time points
corresponding to the last change in the state of the input nodes a,
b, c and can be in the previous or current time-step. The time-step
(time unit) of our model approximately corresponds to nine days.
The randomized asynchronicity of the model does not alter the
steady states of the dynamical system but causes stochasticity in the
trajectory between the initial conditions and the equilibria
(attractors) [32,37], thus it can sample more diverse behaviours
as the traditionally used synchronous models. To determine the
node consensus activity over time (i.e. shared by trajectories with
different update orders) we ran the simulations 100 times and
presented the fraction of simulations in which the node was in an
ON state at a given time-step in the node activity profile. We
confirmed that running the simulations for more than 100 times
did not change the activity profiles.
Our approach of using discrete dynamic modelling allowed us
to sample the timescales of interactions and perform replicate
simulations as well as provide continuously varying activities of the
network nodes over time, which ranged between the lower limit of
0 (below-threshold concentration in all runs) and upper limit of 1
(above-threshold concentration in all runs). However, notice the
exception for IFNc expression higher than one in the helminth
infections. While these activities cannot be directly compared to
quantitative concentrations, we could compare the qualitative
features of the time courses and ask: are they saturating? Do they
show single or multiple peaks? We could also compare the relative
trends of similar variables. It is important to stress that the
empirical data on B. bronchiseptica-T. retortaeformis co-infection were
not used as inputs to the co-infection model but only to validate
the simulated course and intensity of immune responses during co-
infection.
Laboratory experiments
The
primary
single
infections
of
naı¨ve
rabbits
with
B.
bronchiseptica strain RB50 and T. retortaeformis have been described
in detail in Pathak et al. [28] and Murphy et al. [31]. The co-
infection of naı¨ve rabbits with a primary dose of B. bronchiseptica
RB50 and T. retortaeformis followed similar procedures. Here, we
report a concise description of the experimental design, quanti-
fication of the immune variables and parasite intensities.
Ethics statement.
All listed animal procedures were pre-
approved by the Institutional Animal Care and Use Committee of
The Pennsylvania State University.
Co-infection study design.
Out-bred 60 days old New
Zealand White male rabbits were intra-nasally inoculated with
1 ml of PBS solution containing 2.56104 B. bronchiseptica RB50
and simultaneously orally challenged with a 5 ml mineral water
solution of 5,500 infective third stage T. retortaeformis larvae (L3).
Control individuals were treated with 1 ml of PBS or 5 ml of
water, respectively. Groups of 6 individuals (4 infected and 2
controls) were euthanized at days 3, 7, 14, 30, 60, 90, 120 post
challenge and both the respiratory tract and small intestine were
removed to quantify: parasite abundance, cytokine expression in
the lungs and small intestine (duodenum) and mucus-specific anti-
helminth antibody levels (IgA and IgG) from the duodenum to the
ileum (Section SI-1 to SI-4). Blood samples were collected weekly
and used for serum-specific antibody quantification against both
parasites and leukocyte cells count [28,31].
Parasite quantification.
A fixed amount of lungs (15 ml),
trachea (5 ml) and nasal cavity (15 ml), homogenized in PBS, was
serial diluted onto BG blood agar plates supplemented with
streptomycin and incubated at 37uC for 48 hours for bacteria
quantification (Colony forming units, CFU) [28]. The four sections
of the small intestine (SI-1 to SI-4) were washed over a sieve
(100 mm) and helminths collected and stored in 50 ml tubes.
Parasites were counted in five 2.5 ml aliquots and the mean
number, developmental stage and sex (only for adults) estimated in
the four sections [31].
Local cytokine gene expression.
The expression of IFNc,
IL-4 and IL-10 in the lung and duodenum was determined using
Taqman qRT- PCR. RNA isolation, reverse transcription and
qRT-PCR quantification were performed following protocols we
have developed [28,31].
Antibody detection: Antibody IgA and IgG against B. bronchiseptica
and adult T. retortaeformis were quantified using Enzyme-Linked
Immunosorbance Assay (ELISA) [28,31]. Optimal dilutions and
detector antibody against the two parasites were selected by
visually identifying the inflection point from the resulting dilution
curves. For B. bronchiseptica serum dilutions were: 1:10 for IgA and
1:10,000 for IgG, secondary detection antibody: IgA 1:5,000 and
IgG 1:10,000. For T. retortaeformis mucus dilution was: 1:10 both
for IgA and IgG and 1:5,000 for the secondary antibody. We
found cross-reactivity at the antibody level between the somatic
third stage infective larvae (L3) and the adults both in the serum
and the mucus [31]. As such and for simplicity, the empirical data
and the network models were based on the antibody response to
the adult helminth stage.
Haematology.
Blood in anti-coagulated EDTA tubes was
processed using the Hemavet 3 haematology system (Drew
Scientific,
USA)
and
the
general
haematological
profile
quantified [28].
Statistical analysis.
Linear mixed effect models (LME-
REML) were applied to identify changes in the immune
variables during the course of the co-infection and between
single and co-infection. The individual identification code (ID) was
included as a random effect and an autoregressive function of
order 1 (AR-1) was integrated to take into account the non-
independent sampling of the same individual through time or the
monitoring of different parts of the same organ from the same
individual.
To
identify
the
combination
of
immunological
variables that mainly affected parasite abundance a principal
component analysis (PCA singular value decomposition) was used
[31]. Briefly, the strongest linear combination of variables along
the two main PC axes was identified; generalized linear models
(GLM) were then used to examine how parasite abundance was
influenced by each PC axis. To compare the immune variables
between single and co-infection, data from infected animals were
initially scaled over the controls as: Xij* = Xij-Xc, where Xij is an
immune variable for individual i at time j and Xc is the total
average of the controls across the infection for that variable.
Supporting Information
Table S1
Relationship between B. bronchiseptica abundance
(CFU/g) and immune variables from the co-infection experiment.
A- Summary of the Principal Component Analysis (PCA) based on
the most representative immune variables; only the first two PCA
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
17
January 2012 | Volume 8 | Issue 1 | e1002345
axes are reported. Note that the cytokine Ct values are inversely
related to the level of expression. B- Summary of the generalized
linear model (GLM) between bacteria abundance and PCA axis 1
and axis 2.
(DOC)
Table
S2
Relationship between T. retortaeformis abundance
(worm/duodenum length) and immune variables from the co-
infection experiment. A- Summary of the Principal Component
Analysis (PCA) based on the most representative immune
variables; only the first two PCA axes are reported. Note that
the cytokine Ct values are inversely related to the level of
expression. B- Summary of the generalized linear model (GLM)
between helminth abundance and PCA axis 1 and axis 2.
(DOC)
Text S1
Transfer functions for every node of each network: A-
Single B. bronchiseptica infection; B- single T. retortaeformis infection;
C- B. bronchiseptica-T. retortaeformis co-infection. In the functions we
depict the nodes in the intestine with the suffix ‘t’ and the nodes in
the lungs with the suffix ‘b’. Abbreviations: Oag: O-antigen;
IL4II: Interleukin 4 in systemic compartment; DNE: Dead
neutrophils; NE: Recruited neutrophils; IL12I: Interleukin 12 in
lungs/intestine; IgA: Antibody A; C: Complement; TrII: T
regulatory cells in systemic compartment; IL4I: Interleukin 4 in
lungs/small intestine; Th2II: Th2 cells in systemic compartment;
TrI: T regulatory cells in lungs/small intestine; Th2I: Th2 cells in
lungs/small intestine; IL10II: Interleukin 10 in systemic com-
partment; TTSSII: Type three secretion system in systemic
compartment; TTSSI: Type three secretion system in lungs; IgG:
Antibody G; IgE: Antibody E; IL10I: Interleukin 10 in lungs/
small intestine; IFNcII: Interferon gamma in systemic compart-
ment; IFNcI: Interferon gamma in lungs/small intestine; IL12II:
Interleukin 12 in systemic compartment; BC: B cells; DCII:
Dendritic cells in systemic compartment; DCI: Dendritic cells in
lungs/small intestine; Th1I: T helper cells subtype I in lungs/
small intestine; PIC: Pro-inflammatory cytokines; Th1II: T helper
cells subtype I in systemic compartment EC: Epithelial cells lungs/
intestine; AP: Activated phagocytes; T0: Naı¨ve T cells; AgAb:
Antigen-antibody complexes; MP: Macrophages in lungs; EL2:
recruited eosinophils; EL: resident eosinophils; IL13: Interleukin
13; IL5: Interleukin 5; TEL: total eosinophils; TNE: total
neutrophils; TR: T. retortaeformis, Bb: B. bronchiseptica DNE: dead
neutrophils; IS: T. retortaeformis Larvae; AD: T. retortaeformis Adults;
PH: Phagocytosis.
(DOC)
Acknowledgments
We are grateful to Ashley Ruscio for her technical support with the
laboratory co-infection experiment.
Author Contributions
Conceived and designed the experiments: IMC. Performed the experi-
ments: AKP LM IMC. Analyzed the data: JT RA IMC. Contributed
reagents/materials/analysis tools: AKP LM IMC. Wrote the paper: IMC
JT RA. Conceived the immune network models: JT, RA, IMC. Performed
the simulations: JT and RA.
References
1. Cox FE (2001) ‘‘Concomitant infections, parasites and immune responses’’.
Parasitology 122: S23–38.
2. Cattadori IM, Albert R, Boag B (2007) Variation in host susceptibility and
infectiousness generated by co-infection: the myxoma-Trichostrongylus retortaeformis
case in wild rabbits. J R Soc Interface 4: 831–40.
3. Graham AL (2008) Ecological rules governing helminth-microparasite coinfec-
tion. Proc Natl Acad Sci U S A 105: 566–70.
4. Brady MT, O’Neill SM, Dalton JP, Mills KHG (1999) Fasciola hepatica suppresses
a Protective Th1 Response against Bordetella pertussis. Infect Immun 67: 5372–78.
5. Bourke CD, Maizels RM, Mutapi F (2011) Acquired immune heterogeneity and
its sources in human helminth infection. Parasitology 138: 139–59.
6. Anthony RM, Rutitzky LI, Urban JF, Stadecker MJ, Gause WC (2007)
Protective immune mechanisms in helminth infection. Nature Rev Immun 7:
975–87.
7. Allen JE, Maizels RM (2011) Diversity and dialogue in immunity to helminths.
Nature Rev Immun 11: 375–88.
8. Thakar J, Pilione M, Kirimanjeswara G, Harvill E, Albert R (2007) Modeling
systems-level regulation of host immune responses. PLoS Comput Biol 3: e109.
9. Figueiredo CA, Barreto ML, Rodrigues LC, Cooper PJ, Silva NB, et al. (2010)
Chronic intestinal helminth infections are associated with immune hyporespon-
siveness and induction of a regulatory network. Inf Immun 78: 3160–167.
10. Turner JD, Jackson JA, Faulkner H, Behnke J, Else KJ, et al. (2008) Intensity of
Intestinal Infection with Multiple Worm Species Is Related to Regulatory
Cytokine Output and Immune Hyporesponsiveness. J Infect Dis 197: 1204–12.
11. Fox JG, Beck P, Dangler CA, Whary MT, Wang TC, et al. (2000) Concurrent
enteric helminth infection modulates inflammation and gastric immune
responses and reduces helicobacter-induced gastric atrophy. Nature Med 6:
536–42.
12. Chen CC, Louie S, McCormick BA, Walker WA, Shi HN (2006) Helminth-
primed dendritic cells alter the host response to enteric bacterial infection.
J Immunol 176: 472–83.
13. Resende Co T, Hirsch CS, Toossi Z, Dietze R, Ribeiro-Rodrigues R (2006)
Intestinal helminth co-infection has a negative impact on both anti-Mycobac-
terium tuberculosis immunity and clinical response to tuberculosis therapy. Clin
Exp Immunol 147: 45–52.
14. Stewart GR, Boussinesq M, Coulson T, Elson L, Nutman T, et al. (1999)
Onchocerciasis modulates the immune response to mycobacterial antigens. Clin
Exp Immunol 117: 517–23.
15. Haukisalmi V, Henttonen H (1993) Coexistence in Helminths of the Bank Vole
Clethrionomys glareolus II. Intestinal Distribution and Interspecific Interactions.
J Anim Ecol 62: 230–38.
16. Stock TM, Holmes JC (1988) Functional relation-ships and microhabitat
distributions of enteric helminths of grebes (Podicipedidae): the evidence for
interactive communities. J Parasitology 74: 214–27.
17. Khan WI, Blennerhasset PA, Varghese AK, Chowdhury SK, Omsted P, et al.
(2002) Intestinal nematode infection ameliorates experimental colitis in mice.
Infect Immun 70: 5931–37.
18. Wang LJ, Cao Y, Shi HN (2008) Helminth infections and intestinal
inflammation. World J Gastroenterol 14: 5125–32.
19. Holmgren J, Czerkinsky C (2005) Mucosal immunity and vaccines. Nature Med
11: S45–S53.
20. Matzinger P, Kamala T (2011) Tissue-based class control: the other side of
tolerance. Nat Rev Immunol 11: 221–30.
21. Cattadori IM, Boag B, Bjørnstad ON, Cornell S, Hudson PJ (2005) Immuno-
epidemiology and peak shift in a seasonal host-nematode system. Proc R Soc B
272: 1163–69.
22. Pathak AK, Boag B, Poss M, Harvill E, Cattadori IM (2011) Seasonal incidence
of Bordetella Bronchiseptica in an age-structured free-living rabbit population.
Epidem Infect 139: 1210–9.
23. Gueirard P, Weber C, Le Coustumier A, Guiso N (1995) Human Bordetella
bronchiseptica infection related to contact with infected animals: persistence of
bacteria in host. J Clin Microbiol 33: 2002–26.
24. Mattoo S, Cherry JD (2005) Molecular pathogenesis, epidemiology, and clinical
manifestations of respiratory infections due to Bordetella pertussis and other
Bordetella subspecies. Clin Microbiol Rev 18: 326–82.
25. Pilione MR, Harvill ET (2006) The Bordetella bronchiseptica type III secretion
system inhibits gamma interferon production that is required for efficient
antibody-mediated bacterial clearance. Infect Immun 74: 1043–9.
26. Pishko EJ, Kirimanjeswara GS, Pilione MR, Gopinathan L, Kennett MJ, et al.
(2004) Antibody-mediated bacterial clearance from the lower respiratory tract of
mice requires complement component C3. Eur J Immunol 34: 184–93.
27. Kirimanjeswara GS, Mann PB, Harvill ET (2003) Role of antibodies in
immunity to Bordetella infections. Infect Immun 71: 1719–24.
28. Pathak AK, Creppage K, Werner J, Cattadori IM (2010) Immune regulation of
a chronic bacteria infection and consequences for pathogen transmission: the
case of Bordetella bronchiseptica in rabbits. BMC microbiol 10: 226.
29. Audebert F, Hoste H, Durette-Desset MC (2002) Life cycle of Trichostrongylus
retortaeformis in its natural host, the rabbit (Oryctolagus cuniculus). J Helminthol 76:
189–92.
30. Cornell S, Bjørnstad ON, Cattadori IM, Boag B, Hudson PJ (2008) Seasonality,
cohort-dependence and the development of immunity in a natural host-
nematode system. Proc R Soc B 275: 473–591.
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
18
January 2012 | Volume 8 | Issue 1 | e1002345
31. Murphy L, Nalpas N, Stear M, Cattadori IM (2011) Explaining patterns of
infection in free living populations using laboratory immune experiments.
Parasite Immunol 33: 287–302.
32. Assmann SM, Albert R (2009) Discrete dynamic modeling with asynchronous
update, or how to model complex systems in the absence of quantitative
information. Methods Mol Biol 553: 207–25.
33. Glass L, Siegelmann HT (2010) Logical and symbolic analysis of robust
biological dynamics. Curr Opin Genet Dev 20: 644–49.
34. Bornholdt S (2008) Boolean network models of cellular regulation: prospects and
limitations. J R Soc Interface 6: S85–94.
35. Thakar J, Albert R (2010) Boolean models of within-host immune interactions.
Curr Opin Microbiol 13: 377–81.
36. Chaves M, Albert R, Sontag ED (2005) Robustness and fragility of Boolean
models for genetic regulatory networks. J Theor Biol 235: 431–49.
37. Saadatpour A, Albert I, Albert R (2010) Attractor analysis of asynchronous
Boolean models of signal transduction networks. J Theor Biol 266: 641–56.
38. Zhang R, Shah MV, Yang J, Nyland SB, Liu X, et al. (2008) Network model of
survival signaling in large granular lymphocyte leukemia. Proc Natl Acad
Sci U S A 105: 16308–13.
39. Wolfe DN, Kirimanjeswara GS, Goebel EM, Harvill ET (2007) Comparative
role of immunoglobulin A in protective immunity against the Bordetellae. Infect
Immun 75: 4416–22.
40. Abraham DL, Schnyder-Candrian S, Wang CC, Galioto AM, Kerepesi LA,
et al. (2004) Immunoglobulin E and Eosinophil-Dependent Protective Immunity
to Larval Onchocerca volvulus in Mice Immunized with Irradiated Larvae. Infect
Immun 2: 810–817.
41. Brattig NW, Tischendorf FW, Strote G, Medina-de la Garza CE (1991)
Eosinophil-larval-interaction in onchocerciasis: heterogeneity of in vitro
adherence of eosinophils to infective third and fourth stage larvae and
microfilariae of Onchocerca volvulus. Parasite Immunol 13: 13–22.
42. Boyd AP, Ross PJ, Conroy H, Mahon N, Lavelle EC, et al. (2005) Bordetella
pertussis adenylate cyclase toxin modulates innate and adaptive immune
responses: distinct roles for acylation and enzymatic activity in immunomod-
ulation and cell death. J Immunol 175: 730–38.
43. Bancroft AJ, McKenzie AN, Grencis RK (1998) A critical role for IL-13 in
resistance to intestinal nematode infection. J Immunol 160: 3453–61.
44. Finkelman FD, Wynn TA, Donaldson DD, Urban JF (1999) The role of IL-13 in
helminth-induced inflammation and protective immunity against nematode
infections. Curr Opin Immunol 11: 420–26.
45. Pesce JT, Liu Z, Hamed H, Alem F, Whitmire J, et al. (2008) Neutrophils clear
bacteria associated with parasitic nematodes augmenting the development of an
effective Th2-type response. J Immunol 180: 464–74.
46. Harrison GB, Pulford HD, Hein WR, Barber TK, Shaw RJ, et al. (2003)
Immune rejection of Trichostrongylus colubriformis in sheep; a possible role for
intestinal mucus antibody against an L3-specific surface antigen. Parasite
Immunol 25: 45–53.
47. Lacroux C, Nguyen THC, Andreoletti O, Prevot F, Grisez C, et al. (2004)
Haemonchus contortus (Nematoda: Trichostrongylidae) infection in lambs elicits an
unequivocal Th2 immune response. Vet Res 37: 607–22.
48. Henderson NG, Stear MJ (2006) Eosinophil and IgA responses in sheep infected
with Teladorsagia circumcincta Vet Immunol Immunopathol 112: 62–66.
49. Behm CA, Ovington KS (2000) The Role of Eosinophils in Parasitic Helminth
Infections: Insights from Genetically Modified Mice. Parasitol Today 16:
202–09.
50. Jackson JA, Turner JD, Rentoul L, Faulkner H, Behnke JM, et al. (2004) T
helper cell type 2 responsiveness predicts future susceptibility to gastrointestinal
nematodes in human. J Infect Dis 190: 1804–11.
51. Blish CA, Sangare´ L, Herrin BR, Richardson BA, John-Stewart G, et al. (2010)
Changes in plasma cytokines after treatment of Ascaris lumbricoides infection in
individuals with HIV-1 infection. J Infect Dis 201: 1816–21.
52. Geiger SM, Massara CL, Bethony J, Soboslay PT, Carvalho OS, et al. (2002)
Cellular responses and cytokine profiles in Ascaris lumbricoides and Trichuris trichiura
infected patients. Parasite Immunol 24: 499–509.
53. Graham A, Cattadori IM, Lloyd-Smith J, Ferrari M, Bjornstad ON (2007)
Transmission consequences of co-infection: cytokines writ large? Trends in
Parasitol 6: 284–91.
54. Pathak AK, Biares M, Cattadori IM (2011) Snapshot of spatio-temporal cytokine
response to helminth-bacteria single and co-infections. Results in Immunolo-
gy. doi:10.1016/j.rinim.2011.10.001.
55. Xu X, Wen X, Chi Y, He L, Zhou S, et al. (2010) Activation-induced T helper
cell death contributes to Th1/Th2 polarization following murine Schistosoma
japonicum infection. J Biomed Biotechnol. doi:10.1155/2010/202397.
56. Fahel JS, Macedo GC, Pinheiro CS, Caliari MV, Oliveira SC (2010) IPSE/
alpha-1 of Schistosoma mansoni egg induces enlargement of granuloma but does
not alter Th2 balance after infection. Parasite Immunol 32: 345–53.
57. Grencis RK (2001) Cytokine regulation of resistance and susceptibility to
intestinal nematode infection - from host to parasite. Vet Parasitol 100: 45–50.
58. Li S, Assmann SM, Albert R (2006) Predicting essential components of signal
transduction networks: a dynamic model of guard cell abscisic acid signaling.
PLoS Biol 4: e312.
59. Espinosa-Soto C, Padilla-Longoria P, Alvarez-Buylla ER (2004) A gene
regulatory network model for cell-fate determination during Arabidopsis
thaliana flower development that is robust and recovers experimental gene
expression profiles. Plant Cell 16: 2923–39.
60. Sanchez L, Thieffry D (2003) Segmenting the fly embryo: a logical analysis of the
pair-rule cross-regulatory module. J Theor Biol 224: 517–37.
61. Thakar J, Saadatpour A, Harvill E, Albert R (2009) Constraint Based Network
Model of Immune Responses. J R Soc Interface 6: 599–612.
62. Beyer T, Busse M, Hristov K, Gurbiel S, Smida M, et al. (2011) Integrating
Signals from the T-Cell Receptor and the Interleukin-2 Receptor. PLoS Comput
Biol 7: e1002121.
63. Campbell C, Yang S, Albert R, Shea K (2011) A network model for plant–
pollinator community assembly. Proc Natl Acad Sci U S A 108: 197–202.
64. Walsh ER, Thakar J, Stokes K, Huang F, Albert R, et al. (2011) Computational
and experimental analysis reveals a requirement for eosinophil-derived IL-13 for
the development of allergic airway responses in C57BL/6 mice. J Immunol 86:
2936–49.
Immuno-network in Single and Co-infections
PLoS Computational Biology | www.ploscompbiol.org
19
January 2012 | Volume 8 | Issue 1 | e1002345
|
22253585
|
MPI_Bacterium = ( IFNg_Bacterium AND ( ( ( Bb ) ) ) ) OR ( PIC AND ( ( ( Bb ) ) ) )
Th2I_Bacterium = ( Th2II_Bacterium )
Oag = ( Bb )
Th1II_TRetortaeformis = ( DCII_TRetortaeformis AND ( ( ( T0 ) ) AND ( ( DCII_TRetortaeformis ) ) AND ( ( IL12II ) ) ) )
Bb = ( ( Bb ) AND NOT ( PH ) )
PIC = ( ( ( EC_TRetortaeformis ) AND NOT ( IL10I ) ) AND NOT ( IgA_TRetortaeformis ) ) OR ( ( ( EC_Bacterium ) AND NOT ( IL10I ) ) AND NOT ( IgA_TRetortaeformis ) ) OR ( ( ( AD ) AND NOT ( IL10I ) ) AND NOT ( IgA_TRetortaeformis ) ) OR ( ( ( AP ) AND NOT ( IL10I ) ) AND NOT ( IgA_TRetortaeformis ) )
IFNgI = ( Th1I_TRetortaeformis ) OR ( IFNg_Bacterium ) OR ( DCI_TRetortaeformis )
Th2I_TRetortaeformis = ( Th2II_TRetortaeformis )
DCII_Bacterium = ( DCI_Bacterium )
IL12II = ( ( DCII_Bacterium AND ( ( ( T0 ) ) ) ) AND NOT ( IL4II ) ) OR ( ( DCII_TRetortaeformis AND ( ( ( T0 ) ) ) ) AND NOT ( IL4II ) )
EC_Bacterium = ( Bb )
AD = ( ( ( IgG AND ( ( ( IS OR AD ) ) ) ) AND NOT ( MPI_Bacterium ) ) AND NOT ( NE_TRetortaeformis ) )
IL5 = ( Th2II_TRetortaeformis ) OR ( EL2 )
IL10I = ( IL10I_Bacterium ) OR ( Th2I_TRetortaeformis )
IgG = ( BC_TRetortaeformis )
Th1I_TRetortaeformis = ( Th1II_TRetortaeformis )
NE_Bacterium = ( PIC )
IL4I = ( IL4II )
Th2II_TRetortaeformis = ( ( DCII_TRetortaeformis AND ( ( ( T0 ) ) ) ) AND NOT ( IL12II ) )
DP = ( NE_Bacterium AND ( ( ( TTSSI ) ) ) )
AP = ( AgAb_Bacterium AND ( ( ( Th1I_Bacterium AND MPI_Bacterium ) ) AND ( ( Bb ) ) ) ) OR ( IgG_Bacterium AND ( ( ( Cb ) ) AND ( ( Th1I_Bacterium AND MPI_Bacterium ) ) AND ( ( Bb ) ) ) )
TTSSI = ( ( ( Bb ) AND NOT ( IgA_Bacterium ) ) AND NOT ( IgG_Bacterium ) )
DCI_Bacterium = ( IFNg_Bacterium AND ( ( ( Bb ) ) ) ) OR ( PIC AND ( ( ( Bb ) ) ) )
IL4II = ( EL2 ) OR ( ( ( Th2II_Bacterium ) AND NOT ( IL12II ) ) AND NOT ( IFNgI ) ) OR ( ( ( Th2II_TRetortaeformis ) AND NOT ( IL12II ) ) AND NOT ( IFNgI ) ) OR ( ( ( DCII_Bacterium AND ( ( ( T0 ) ) ) ) AND NOT ( IL12II ) ) AND NOT ( IFNgI ) ) OR ( ( ( DCII_TRetortaeformis AND ( ( ( T0 ) ) ) ) AND NOT ( IL12II ) ) AND NOT ( IFNgI ) )
EL2 = ( IgE AND ( ( ( IL5 ) ) ) ) OR ( IL13 AND ( ( ( IL5 ) ) ) )
IgE = ( IL4II AND ( ( ( BC_TRetortaeformis ) ) ) ) OR ( IL13 AND ( ( ( BC_TRetortaeformis ) ) ) )
TTSSII = ( TTSSI )
TNE = ( NE_TRetortaeformis ) OR ( NE_Bacterium )
IFNg_Bacterium = ( DCI_Bacterium ) OR ( MPI_Bacterium ) OR ( ( ( Th1I_Bacterium ) AND NOT ( IL10I_Bacterium ) ) AND NOT ( IL4I ) )
BC_Bacterium = ( T0 ) OR ( BC_Bacterium )
EL = ( ( IS ) AND NOT ( EL2 ) )
IL10I_Bacterium = ( TrI_Bacterium ) OR ( Th2I_Bacterium AND ( ( ( TTSSI ) ) ) ) OR ( MPI_Bacterium )
T0 = ( DCII_Bacterium ) OR ( DCII_TRetortaeformis )
Th2II_Bacterium = ( ( DCII_Bacterium AND ( ( ( T0 ) ) ) ) AND NOT ( IL12II ) )
IFNgII = ( IFNg_Bacterium ) OR ( IFNgI )
EC_TRetortaeformis = ( IS ) OR ( AD )
Th1II_Bacterium = ( DCII_Bacterium AND ( ( ( T0 AND IL12II ) ) ) )
PH = ( AP AND ( ( ( Bb ) ) ) )
DCI_TRetortaeformis = ( PIC )
Cb = ( AgAb_Bacterium AND ( ( ( IgG_Bacterium ) ) ) ) OR ( ( Bb ) AND NOT ( Oag ) )
IgA_Bacterium = ( BC_Bacterium AND ( ( ( Bb ) ) ) ) OR ( IgA_Bacterium AND ( ( ( Bb ) ) ) )
BC_TRetortaeformis = ( BC_TRetortaeformis ) OR ( T0 )
Th1I_Bacterium = ( Th1II_Bacterium )
TrI_Bacterium = ( TrII )
IL13 = ( Th2I_Bacterium ) OR ( Th2I_TRetortaeformis ) OR ( EL2 ) OR ( EL AND ( ( ( IS ) ) ) )
IgG_Bacterium = ( BC_Bacterium ) OR ( IgG_Bacterium )
DCII_TRetortaeformis = ( DCI_TRetortaeformis )
AgAb_Bacterium = ( IgA_Bacterium AND ( ( ( Bb ) ) ) ) OR ( IgG_Bacterium AND ( ( ( Bb ) ) ) )
NE_TRetortaeformis = ( ( ( IFNgI ) AND NOT ( IL4I ) ) AND NOT ( IL10I ) ) OR ( PIC AND ( ( ( AD ) ) ) )
TEL = ( EL2 ) OR ( EL )
TrII = ( DCII_Bacterium AND ( ( ( TTSSII ) ) AND ( ( T0 ) ) ) )
IgA_TRetortaeformis = ( IS AND ( ( ( BC_TRetortaeformis ) ) ) )
|
Helikar et al. BMC Systems Biology 2012, 6:96
http://www.biomedcentral.com/1752-0509/6/96
SOFTWARE
Open Access
The Cell Collective: Toward an open and
collaborative approach to systems biology
Tom´aˇs Helikar1*, Bryan Kowal2, Sean McClenathan2, Mitchell Bruckner1, Thaine Rowley1,
Alex Madrahimov1, Ben Wicks2, Manish Shrestha2, Kahani Limbu2 and Jim A Rogers1,3
Abstract
Background: Despite decades of new discoveries in biomedical research, the overwhelming complexity of cells has
been a significant barrier to a fundamental understanding of how cells work as a whole. As such, the holistic study of
biochemical pathways requires computer modeling. Due to the complexity of cells, it is not feasible for one person or
group to model the cell in its entirety.
Results: The Cell Collective is a platform that allows the world-wide scientific community to create these models
collectively. Its interface enables users to build and use models without specifying any mathematical equations or
computer code - addressing one of the major hurdles with computational research. In addition, this platform allows
scientists to simulate and analyze the models in real-time on the web, including the ability to simulate loss/gain of
function and test what-if scenarios in real time.
Conclusions: The Cell Collective is a web-based platform that enables laboratory scientists from across the globe to
collaboratively build large-scale models of various biological processes, and simulate/analyze them in real time. In this
manuscript, we show examples of its application to a large-scale model of signal transduction.
Background
The immense complexity in biological structures and pro-
cesses such as intracellular signal transduction networks
is one of the obstacles to fully understanding how these
systems function. As understanding of these biochemical
pathways increases, it is clear that they form networks of
astonishing complexity and diversity. This means that the
complex pathways involved in regulation of one area of the
cell (so complex that a researcher could spend their entire
career working in that area alone) are so interconnected to
other, equally complex areas that all of the different path-
way systems must be studied together, as a whole, if any
of the individual components are to be understood. How-
ever, the large scale and minute intricacy of each of the
individual networks makes it difficult for cell biologists or
biochemists working in one area of a cell’s biochemistry
to be aware of, let alone relate their results to, findings
obtained from the various different areas. So how will all
*Correspondence: thelikar@unomaha.edu
1Department of Mathematics, University of Nebraska at Omaha, Omaha, NE,
USA
Full list of author information is available at the end of the article
of these individually complex systems be possible to study
in an integrated biochemical “mega-system?”
In order to address this problem, the concept of systems
biology study has emerged [1-8]. However, with i) data
being generated by laboratory scientists at a staggering
rate in the course of studying the individual systems, ii)
the fact that these individual systems are so complicated
that scientists rarely have detailed knowledge about areas
outside those that they study, there is a huge imped-
iment to implementing a systems approach in cellular
biochemistry, and iii) for laboratory scientists to fully
embrace systems biology computational tools must lend
themselves to usage without requiring advanced mathe-
matical entry or programming.
Several significant advancements in the systems biology
field have been made as a response to the sea of data
being generated at ever increasing rates. For example,
in the area of biochemical signal transduction, several
community-based projects to organize information about
signal transduction systems such as the Alliance for Cellu-
lar Signaling [9], the former Signal Transduction Knowl-
edge Environment [10], UniProt [11], or the WikiPathways
project [12] have been created. These resources provide a
© 2012 Helikar et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly cited.
Helikar et al. BMC Systems Biology 2012, 6:96
Page 2 of 14
http://www.biomedcentral.com/1752-0509/6/96
way to organize and store important laboratory-generated
data and information such as gene sequences, protein
characteristics, interaction partners, etc.; these are then
easily accessible via the Internet to the scientific commu-
nity. Building on these resources and advancements has
been the development of tools to visualize and analyze
these data and, specifically, the entities that make up the
complex, network-like structures of biological processes.
Amongst the most widely used tools to visualize biological
networks is the open-source software, Cytoscape [13].
The information contained in the above database
resources (and visualized via Cytoscape) is limited in
that it is mostly static; biological systems however are
dynamic in nature. Hence to fully understand the under-
lying mechanisms (and those of corresponding diseases),
the dynamics of these processes need to be considered.
Computational modeling and simulation has been suc-
cessfully adopted in a number of fields to dramatically
reduce development costs. The use of these modern tools
to organize and probe biological structure and function
has a high potential to provide the basis for new break-
throughs in both basic understanding of cell function
and the development of disease therapies. The ability
to observe the actual dynamics of large scale biolog-
ical systems increases the probability that, out of the
tens of thousands of combinations of interactions, unex-
pected points of intervention might be deciphered. The
Cell Collective aims at providing an environment and
resource where the biomedical community, as a whole,
can more effectively bring these exciting new computa-
tional approaches to bear on cellular systems. The inte-
gration of computational and laboratory research has the
potential to lead to improved understanding of biological
processes, mechanisms of disease, and drug development.
If a “systems approach” is to be successful, then there
must be a “system” into which the thousands of labora-
tory scientists all over the world can incorporate their
detailed local knowledge of the pathways to create a global
model of biochemical pathways. With such a systems plat-
form, all local information would be far more accurate
if laboratory scientists would contribute their specialized
expertise into a system that enables the integration of
the currently dispersed knowledge. Hence, a collabora-
tive modeling platform has the potential to substantially
impact and move forward biomedical research.
This is precisely the purpose of The Cell Collective.
The Cell Collective is an environment to model biolog-
ical processes. The platform allows scientists to deposit
and track dynamical information about biological pro-
cesses and integrate and interrogate this knowledge in the
context of the biological process as a whole. Laboratory
scientists can directly simulate large-scale models in real
time to not only help test and form new hypotheses for
their laboratory research, but also to make research more
easily reproducible (through sharing their models with
collaborators). Furthermore, the creation and simulation
of models in The Cell Collective doesn’t require direct use
of mathematics or programming – a substantial advance-
ment in the field [14]; this tool has been developed to
bring modeling into the hands of mainstream laboratory
scientists.
The role of The Cell Collective in the current landscape of
systems biology technology
As a result of the constant flow of data from laborato-
ries, the success of biomedical research relies now, more
than ever, on computational and computer technologies.
While a number of different technologies have already
been developed and succeeded in their purpose, The Cell
Collective further builds on the successes of these efforts
to provide a novel technology to exploit the full potential
of systems biology. In this section, a discussion of some of
these technologies follows. Note that, the following is not
an extensive review, rather we aim to illustrate how The
Cell Collective fits within the landscape of systems biology
resources. For better understanding, these resources have
been categorized according to their function.
A) Biological databases (as mentioned in the Background
section, Alliance for Cellular Signaling [9], STKE
[10], UniProt [11], the WikiPathways project [12],
KEGG [15], UniProt [16], Reactome [17], Pathway
Commons [18], etc.) were developed as one of the
first steps to deal with the sea of biological data being
produced with high-throughput technologies. The
information contained in these biological databases
focuses on static cell “parts lists.” In other words, the
data focuses on the description of the individual
entities rather than the dynamical relationship
between the individual parts. Conversely, The Cell
Collective, and specifically its Knowledge Base
component (discussed in the Results section) extends
static knowledge and data into dynamical models;
hence the information contained in the Knowledge
Base (which is purely qualitative) is dynamical in
nature; it takes into account the dynamical
relationship between all of the interacting partners.
B) Software for dynamical models (which employ
mathematical frameworks similar to the ones used in
The Cell Collective – i.e., rule-based formalisms) also
already exist (e.g., GINsim [19], BooleanNet [20],
CellNetOptimizer [21], or BoolNet [22]). These tools
have been built and used mainly for individual groups
to study networks of a confined size. They also rely on
the users’ training in computer programming and/or
mathematics (and hence are first and foremost tools
developed for modelers); this makes it difficult for
laboratory scientists to incorporate these tools into
Helikar et al. BMC Systems Biology 2012, 6:96
Page 3 of 14
http://www.biomedcentral.com/1752-0509/6/96
their experimental studies. The Cell Collective
provides a novel tool in the area of large-scale, whole
cell models, while extending the use of
computational modeling to laboratory scientists.
C) Model repositories such as the CellML repository
[23] or the BioModels Database provide a central
location to store models developed by the
community. These models are then available to
others for download and further analyses using other
tools. The BioModels Database is primarily a model
repository, however, it does provide simulation
capabilities via the JWS simulator [24]. In addition,
the PathCase systems biology tool [25,26] provides a
central place for kinetic models from the BioModels
Database and KEGG pathways to be queried,
visualized, and simulated side-by-side. Similar to
these resources, The Cell Collective provides the first
repository (with simulation capabilities) for models
based on a qualitative mathematical
formalism.
D) Model exchange standards such as the Systems
Biology Markup Language (SBML, [27,28]) or
CellML [29] make it easier for models to be
exchanged between different groups and
simulated/analyzed by different simulation tools. For
example, when a research group wants to simulate a
model deposited to the BioModels Database, the
model’s description in SBML or CellML ensures that
the model truly corresponds to the same model used
by a different group, and hence the generated data
can be easily reproduced. While users can share their
models with other users of The Cell Collective
directly, without the need to import/export model
files, the platform currently provides SBML export
features based on the most recent version of SBML
L3 qualitative package [30].
E) Visualization and analysis tools for static interaction
networks, such as the aforementioned Cytoscape
[13], but also others including VisANT [31] or Gephi
(http://gephi.org), have been used extensively to
visualize and analyze the graph properties of
networks of various types and sizes. As a
complement to existing graph analyses, The Cell
Collective deals with dynamical models – ones that
can be put in motion via computer simulations – and
hence focuses on the visualization of the dynamics of
these models via simulations, and susbsequent
analyses (e.g., input-output relationships). Together,
The Cell Collective is a platform that not only
provides a unique combination of successful systems
biology and modeling approaches, but also offers
significant innovations to these technologies. In this
manuscript, discussed are the various components
and features of the platform, and exemplified on a
previously published large-scale network model of
signal transduction [32].
Implementation
The Cell Collective is a server-based software imple-
mented in Java and powered by MySQL database. The
simulation engine is based on ChemChains which was
implemented in C++ [33]. The user interface of The Cell
Collective was implemented primarily using JavaServer
Faces (http://www.javaserverfaces.org) and Primefaces
(http://www.primefaces.org).
Computational framework and simulations
Models in The Cell Collective are based on a qualita-
tive, rule-based mathematical framework. In this frame-
work, each species can assume either an active or inac-
tive state. Which state a species assumes at any given
time point depends on a set of rules that take into
account the activation state of all immediate upstream
regulators.
The Bio-Logic Builder provides the user interface for
users to enter qualitative information about the regula-
tory mechanism of each species in a model, and sub-
sequently converts this information into an appropriate
mathematical (algebraic) expression (manuscript submit-
ted). Before the simulation engine (ChemChains) can
simulate a model, the mathematical expressions of indi-
vidual species are converted into C++ (.cpp) files, which
are subsequently compiled into a single dynamical library
(.so file). This dynamical library encodes the entire model
which is subsequently simulated by ChemChains (see
Figure 1).
Though
a
discrete
(active/inactive)
mathematical
framework is used to represent the modeled biological
processes, ChemChains has been developed to enable
simulations of discrete models while using continuous
input/output data. In general, the activity levels of the
models’ individual constituents is measured as %ON.
Depending on the context of the biological process being
simulated, this measure corresponds, for example, to
concentration or the fraction of biological species being
active at any given time.
In the case of real-time simulations, %ON of a species
represents its moving average activity, and is calculated
as the fraction of the active/inactive states over a slid-
ing window. For simulations using the Dynamical Analysis
feature, the activity levels of the individual species (or
%ON) also corresponds to the ratio of active/inactive
states, but is calculated once the dynamics of the model
settle in a steady behavior (or an attractor as described
in great detail in [33]). In both the real time simula-
tions and dynamical analysis, %ON is used as a semi-
quantitative way to measure the dynamics of the modeled
biological processes.
Helikar et al. BMC Systems Biology 2012, 6:96
Page 4 of 14
http://www.biomedcentral.com/1752-0509/6/96
Figure 1 Construction of models prior to their simulations via
built-in ChemChains. The bio-logic for each species (defined by
users) is converted (automatically) to a mathematical (Boolean)
expression. Each species’ expression is encoded to a C++ file, and all
files are subsequently compiled into a single dynamic library (.so file)
which can be read and executed by ChemChains for simulations.
Simulation performace
We analyzed the perfomance of individual simulations for
randomly generated models of different sizes and different
complexities (in terms of network connectivity). Specifi-
cally, we considered models with 10, 100, 500, and 1,000
nodes and network connectivities of 2, 5, 10, 20, and
100. Note that for biological application, relatively small
(low single digit) connectivity is most realistic [32,34,35].
As can be seen Table 1, simulations in The Cell Collec-
tive are relatively efficient as the required computational
resources are in a linear relationship with the increasing
parameters of the generated networks.
Results and discussion
The Cell Collective is a web-based platform (accessible at
http://www.thecellcollective.org) in which laboratory sci-
entists can collaboratively build mathematical models of
Table 1 Simulation performance for models with ranging
complexity
# of nodes/
connectivity
2
5
10
20
100
10
0.88s
0.94s
0.85s
0.99s
0.93s
100
4.82s
4.98s
5.55s
5.95s
9.8s
500
26.99s
29.42s
32.11s
37.31s
68.73s
1,000
60.89s
64.61s
70.95s
79.59s
149.34s
Simulations consisted of 10,000 time steps and were performed on a computer
with a single core, 2GHz processor and 2GB of RAM.
biological processes by utilizing existing laboratory data,
and subsequently simulate the models to further guide
their laboratory experiments. Conceptually, the platform
can be broken up into three parts (Figure 2) that form the
basis for the core functionality of the software: 1) inte-
grated Knowledge Base of protein dynamics generated
from laboratory research in a single repository, 2) integra-
tion of this knowledge into mathematical representation
that allows visualization of the dynamics of the data (i.e.,
put it in motion via simulations), and 3) simulations and
analyses of the model dynamics. As can also be seen in
the figure, these three parts form a loop that is closed by
laboratory experimentation. The first model in The Cell
Collective (available in for all users to simulate and build
upon) is one of the largest models of intracellular signal
transduction [32]. Features available in the current version
of The Cell Collective are described in more detail in the
following sections.
Knowledge Base of interaction dynamics
When laboratory scientists produce new results, for
example regarding the role of one protein interacting with
another protein, these results are usually published along
with thousands of other results generated by the scien-
tific community. The publication of individual results in
isolation means that separate findings are not necessarily
absorbed, verified, analyzed, and integrated into the
existing knowledge. With the invention of various high-
throughput technologies, the gap between the amount
of knowledge produced and the ability of the scientific
community to fully utilize this knowledge has grown [36].
The first major component of The Cell Collective (as
highlighted in Figure 2) is a Knowledge Base which
enables laboratory scientists to contribute to the integra-
tion of knowledge about individual biological processes
at the most local level which includes, for example, the
identification of direct protein-protein interactions. How-
ever, the goal of The Cell Collective is not to duplicate
other well-established resources by providing extensive
parts lists that make up various biological processes and
cells. Instead, the aim of the platform is to extend static
knowledge and data into dynamical models; hence the
information provided in the Knowledge Base needs to
be dynamical in nature. This means that the information
(which is purely qualitative – see the Methods section)
contained in The Cell Collective Knowledge Base takes
into account the dynamical relationship between all of the
interacting partners. For example, let’s assume, there are
two positive regulators (X and Y) of a hypothetical species
Z. While in the context of a parts list, information about
the above species and interactions would be sufficient,
in order to abstract the biological process to a dynami-
cal model, one needs to know the dynamical relationship
between the interacting partners. For instance, are both X
Helikar et al. BMC Systems Biology 2012, 6:96
Page 5 of 14
http://www.biomedcentral.com/1752-0509/6/96
Figure 2 Overview of the flow of knowledge about biological processes, and the role of The Cell Collective in integrating and
understanding this knowledge in the context of the biological processes as a whole.
and Y necessary for the activation, or is either one of them
sufficient to activate Z? This is the type of information
that is used to construct dynamical models in The Cell
Collective.
Based on a widely known wiki-like concept, the Knowl-
edge Base module of the platform was developed to
allow laboratory scientists to contribute – collabora-
tively – their knowledge to the complete regulatory
mechanisms of individual biological species. Because all
of the regulatory information forms the basis of the
modeled biological/biochemical process, and hence has
to be correct for the model to exhibit similar behav-
iors as seen in the laboratory, this process of aggre-
gating all known information about a species into one
place can also serve as a mechanism to identify pos-
sible contradictions or holes in the current knowledge
about the regulatory mechanism of a particular species.
Using the previous hypothetical example, let’s assume
laboratory scientist A discovers that proteins X and Y
are both necessary to activate species Z, but scientist
B’s laboratory results suggest either protein X or Y can
sufficiently activate Z (Figure 3). The process of inte-
grating all known information on species Z becomes
crucial in discovering such discrepancies (or additional
missing information), which may have not been found
otherwise. Because the goal of The Cell Collective is
to also integrate this information into dynamical mod-
els, simulations of the large-scale model (which might
have hundreds or thousands of additional components
in it) can suggest whose data is more likely to be
correct. Assume that scientist A adds his informa-
tion into the model and the model exhibits phenom-
ena similar to the ones seen in the laboratory, whereas
when the model is built with the data from scientist
B’s experiments, the simulation dynamics of the over-
all model fails to resemble the known actions of the
real system. In such a case, new laboratory experiments
would be warranted, with a potential to produce more
insights into the regulatory mechanism of protein Z
(Figure 3).
The sea of biological information has made it dif-
ficult for the data to be verified on such an inte-
grated basis. We fully understand how some of the
most complex biological systems work only when the
experimental data is re-integrated into and seen in
the context of the entire system; a platform for inte-
gration of data is exactly what The Cell Collective
provides.
Dynamical information
Each species in The Cell Collective’s Knowledge Base has
a dedicated page where laboratory scientists can directly
deposit their knowledge regarding the species’ regulatory
mechanisms. While the wiki-like format of the Knowl-
edge Base gives users the ability to input their data in
a free form which can be also interactively discussed,
each page is structured to help users organize and review
their data more efficiently. Because the wiki format is
Helikar et al. BMC Systems Biology 2012, 6:96
Page 6 of 14
http://www.biomedcentral.com/1752-0509/6/96
Figure 3 Integration of laboratory results via modeling. The different relationships between hypothetical interactions of X and Y with Z as
discovered by scientists A and B. Solid lines depict the necessity of the interaction for species Z to be activated, whereas dashed lines correspond
the optional nature of the interaction. Because scientist B’s results suggest an “OR” relationship between the regulators, there are two graphical
representations of Z’s regulatory mechanism.
an easy medium for collecting knowledge from a large
number of individuals, a number of scientific efforts have
successfully adopted a variation of this technology (e.g.,
[12][32][33][34]).
First, the Regulation Mechanism Summary section
describes
the
general
mechanism
of
the
activa-
tion/deactivation of the species. This section, found at
the top of the page of a given species, is most important
from a systems perspective as the information therein
takes into an account the role of all immediate upstream
regulators (see below).
The Upstream Regulators section contains the list of key
players that have a role in the regulation of the species,
as well as any evidence (as found in the laboratory) sup-
porting those roles. Using the earlier example involving
the regulatory mechanism of species Z, this section would
include proteins X and Y as upstream regulators, and
the findings of laboratory scientists A and B suggest-
ing the role of these regulators in the activation of the
species (Figure 4). On the other hand, the Regulation
Mechanism Summary section (discussed above) would
contain the overall dynamical information as to how Z
is regulated in the context of both X and Y (i.e., are
both regulators required for the activation, or only one of
them?).
Model-specific Information section: Because a number
of molecular species can be regulated differently based
on the type of the cell, this section allows users to
enter such cell type-specific information. For example,
an intracellular species can be regulated either by dif-
ferent players, or the same players but with different
dynamical relationships in, say, a T cell and a mammary
epithelial cell. This section enables users to differenti-
ate between the regulatory mechanisms of the species
in the two (or more) different types of cells (i.e., mod-
els). Hence, this section can be utilized by users to
define upstream regulators and the regulation mecha-
nism summary that is specific to users’ different mod-
els. For example, the regulation mechanism summary of
species Z in scientist A’s model would describe his find-
ings that both upstream regulators of Z are necessary
for its activation, whereas scientist B’s regulation mech-
anism summary on wiki page for Z would indicate that
either one of the upstream regulators can activate Z
(Figure 4).
Finally, References is a section that users can use to
record any published works that support information
entered in any of the above sections. Users can enter ref-
erences by simply entering the Pubmed ID (pmid) of the
article of interest and The Cell Collective will automati-
cally import all of the bibliographical information about
the works.
As a starting point, we have deposited all biological
knowledge describing one of the largest dynamical models
of signal transduction built and published as part of our
previous research [32]. This model consists of around 400
biochemical interactions between 130 species, comprising
a number of main signaling pathways such as the Epi-
dermal Growth Factor, Integrin, and G-Protein Coupled
Receptor pathways. The dynamical information about
the hundreds of local interactions, collected manually
from published biochemical literature, is available in the
Knowledge Base module. Expert scientists in the field
may begin contributing to it, as well as discovering
Helikar et al. BMC Systems Biology 2012, 6:96
Page 7 of 14
http://www.biomedcentral.com/1752-0509/6/96
Figure 4 Visualization of the flow of data generated by
laboratory scientists through The Cell Collective Knowledge
Base and Bio-Logic Builder. For example scientists A and B identify
different upstream regulators (protein X and Y, respectively) of
protein Z. This knowledge is subsequently recorded in the Upstream
Regulators section on the page of protein Z. Then both scientists A
and B determine what the relationship is between the two upstream
regulators of Z. Once the overall regulation mechanism is agreed
upon, the scientists use Bio-Logic Builder to add the regulatory
mechanism of Z to an actual model. The mathematical representation
of the species bio-logic is generated in the background, so the user
never has to define any mathematical equations nor expressions.
discrepancies and gaps in the biological knowledge that
might have been included in the model.
Once the dynamical information about the individual
interactions is added in the platform Knowledge Base,
the next step is to convert this knowledge into a dynam-
ical model; a discussion on where this piece fits into the
overall concept of The Cell Collective follows in the next
section.
Building computational models
While the Knowledge Base component of The Cell Collec-
tive serves as the knowledge aggregator for the dynamical
regulatory mechanisms of individual biological species,
the next step (#2 in Figure 2) is to convert this knowl-
edge into a dynamical computational model that can be
simulated and analyzed on the computer.
Perhaps one of the biggest challenges in transforming
biological knowledge into a computational model is the
conceptual gap between the mathematical and biological
sciences. Thus far, the creation of mathematical models
has been limited to scientists who are well versed in com-
puter science and mathematics. To address this issue, we
have developed Bio-Logic Builder (manuscript submit-
ted), a component of The Cell Collective, which allows
laboratory scientists to build computational models based
purely on the logic of the species’ regulatory mechanisms
as discovered in the laboratory.
The step of transforming biological knowledge into its
model representation is aided by the information pro-
vided in the Knowledge Base component of the software
platform (Figure 4). Specifically, as discussed above, the
information recorded for the corresponding local interac-
tions by individual scientists amounts to the overall regu-
lation mechanism which represents the blueprint of each
species’ bio-logic. While the local interactions (concerning
a hypothetical protein Z in Figure 4) are discovered in the
laboratory by individual scientists (for example scientists
A and B as shown in the figure), the species overall reg-
ulation mechanism should take into an account all of the
local knowledge (and hence should be determined in a
collaborative fashion). Bio-Logic Builder was developed in
such a way that all that is necessary to construct the com-
putational representation of the regulatory mechanism
of each species is the same qualitative data provided in
the Knowledge Base component. Scientists define each
species’ bio-logic in a modular fashion by simply defin-
ing activators and inhibitors (i.e., upstream regulators)
of the species of interest, as well as the logical relation-
ship between the upstream regulators (e.g., whether or
not a set of activators is required for activation, as dis-
cussed in an example above). Because models in The
Cell Collective utilize a qualitative, rule-based mathemat-
ical framework, no kinetic parameters are necessary to
construct the models. (A quick tutorial on how to use
the Bio-Logic Builder to construct models is available at
http://www.thecellcollective.org)
Once the bio-logic is defined for all species in a
given model, in silico simulations and analyses can be
conducted (step #3 in Figure 2). How this can be
done with The Cell Collective is the focus of the next
section.
Simulations and analyses of model dynamics
The idea behind abstracting biological processes as com-
putational models is to be able to visualize the dynamics
of these processes on the computer, and to conduct in
silico experiments that can provide i) new insights into
laboratory experiments and ii) additional basis for the-
oretical computational research to further elucidate the
complexity governing these biological processes. With its
simulation and analysis component, The Cell Collective
has been designed to provide exactly these features.
Helikar et al. BMC Systems Biology 2012, 6:96
Page 8 of 14
http://www.biomedcentral.com/1752-0509/6/96
Specifically, in the current version of the platform, two
tools for simulations and analyses (discussed below) are
available.
Real-time simulations
Perhaps the most unique and novel innovation to com-
putational modeling is the real-time simulation feature in
the platform, which allows users to visualize the dynamics
of any model interactively and in real time. Similar to the
rest of the platform, the simulation features have been
designed with simplicity and intuitiveness in mind.
All modeled biological/biochemical processes in The
Cell Collective, represented by species that make up the
internal machinery of the cell, are simulated in exter-
nal environments which drive the dynamics of the sys-
tem. In our example of signal transduction, this environ-
ment is represented by external species corresponding to
various extracellular signals such as growth hormones,
stress, etc. Using a simple slider, users can change the
amount of each extracellular signal (measured in %ON
on a scale of 0 to 100 – see the Methods section for
more detail) and visualize the effects of the changes on
the dynamics of the cell while the simulation is running.
Similarly, users can introduce biological mutations to sim-
ulate loss-of-function and gain-of-function experiments
while watching the dynamics of the cell change as a result
of the mutations. For users’ convenience, real time sim-
ulations can be also paused and resumed at any time.
Figure 5 shows a screen-shot of the real time simulation
tool. A short video demonstration of real time simulations
using the previously mentioned large-scale model of sig-
nal transduction is also available as a Additional file 1.
Dynamic Analysis
Laboratory studies to identify functional relationships
between extracellular stimuli and various components of
the cell involve a number of experiments that can be both
time consuming and resource demanding. For example,
a laboratory study [37] that suggests that Akt (a ser-
ine/threonine kinase involved in the regulation of a variety
of cellular responses such as apoptosis, proliferation, etc.)
is activated in response to the Epidermal Growth Factor
Figure 5 Screen-shot of a real-time simulation. Users can change the activity level of the extracellular species via simple sliders (boxed in red).
Each tracing in the graph corresponds to an activity level of a species specified in the legend by the user. Any effects of the change of activity of the
external species is then reflected in the dynamics of the species’ graph; as the user moves the slider, the activity patterns of the selected species
change in real time. In addition, by using the “Mutate” button, users can simulate the effects of gain/loss-of-function mutations on the dynamics the
modeled biological process.
Helikar et al. BMC Systems Biology 2012, 6:96
Page 9 of 14
http://www.biomedcentral.com/1752-0509/6/96
(EGF), the activity of Akt is measured and compared in
untreated cells and cells treated with EGF. Such studies
usually involve the construction of a number of protein
constructs, cell cultures, assays, etc, amounting to the use
of many resources.
While Akt has been known for many years to be
activated in response to EGF, there are many areas of
the cell that are not as well understood. Laboratory
experiments in such areas can be sometimes based on
less sound hypotheses that may lead to the waste of many
resources. But what if one had the ability to pre-test
laboratory hypotheses on the computer, using a com-
putational model, in a matter of minutes? This would
allow laboratory scientists to weed out weak hypotheses
while focusing on the ones that have a better chance of
being proven correct, and hence resulting in more efficient
studies.
This is where the Dynamic Analysis simulation fea-
ture of The Cell Collective plays an important role. This
tool allows users to conduct in silico experiments that
closely resemble the way laboratory experiments are per-
formed, with the advantage that in these computational
studies researchers can perform more simulations and
experiments in a much shorter time-frame. For example,
models in The Cell Collective can be simulated and their
dynamics visualized and analyzed in hundreds or thou-
sands of extracellular environments (as opposed to the
limited number of scenarios possible in the laboratory) in
a manner of minutes.
As an example, we will demonstrate how the soft-
ware can be used to study the relationship between EGF
and Akt. The dynamical analysis studies are done in
two parts. First, on the main page of the simulation
tool (Figure 6), users define the extracellular environ-
ment under which the study will be done. This is anal-
ogous to the preparation of cell media in the laboratory.
Similar to laboratory experiments with real cells, differ-
ent studies using computational models (or virtual cells)
Figure 6 Dynamical analysis page. Dynamical analysis page. For each in silico experiment, users can use the dual sliders to define the ranges of
activity levels of each extracellular species. Users can also set additional properties of the experiment including the number of simulations as well as
mutations (gain/loss-of-function).
Helikar et al. BMC Systems Biology 2012, 6:96
Page 10 of 14
http://www.biomedcentral.com/1752-0509/6/96
also require the set up of optimal extracellular condi-
tions. As visualized in the figure, this can be done easily
by setting the ranges of the activity (from 0 to 100%)
of the individual extracellular (external) species via the
dual sliders (or by just typing the activity levels in the
appropriate text boxes). Because in this example exper-
iment, we are interested in the effects of EGF on the
network model, the activity of EGF (boxed in red) is
set to range on the full scale between 0 and 100% ON.
On the other hand, the activity ranges of the remaining
external species are selected for optimal results based on
our previous research [32], and supported by laboratory-
generated data. For example, the Extracellular Matrix
(ECM) is set to higher activity levels, varying between 56
and 100% (boxed in blue); this corresponds to a biological
finding that EGF-induced growth (as well as other cellu-
lar processes) is dependent on cell anchorage via ECM
[38]. (Note that, from our experience with large-scale
models, while optimal conditions should be determined,
the simulations and results are not sensitive to exact
values.)
While in this example, 100 simulations are performed,
users can specify the number of simulations to be run
within the study (Figure 6). During each simulation, an
activity level for each extracellular species is selected ran-
domly by the software such that the activity falls into the
specified range. As a result, the user is able to simulate
what would amount to 100 different laboratory experi-
ments, with each experiment corresponding to a different
external condition.
Once the in silico experiment has completed, users
can analyze the dynamics of the model. Currently, the
Dynamic Analysis tool allows users to generate dose-
response curves to investigate qualitative (input-output)
relationships between external cellular signals and vari-
ous components of the model, such as the one between
EGF and Akt as visualized in Figure 7. As can be seen in
the graph, there is indeed a positive correlation between
EGF and Akt, similar to the phenomenon seen in the
laboratory. An additional significant advantage of com-
putational experiments using this tool is that users can
generate a number of analyses without re-running the
entire experiment. For instance, in addition to examining
the functional relationship of Akt and growth, one can
generate similar dose-response curves for any species in
the model using a single 100-simulation experiment. This
Figure 7 An example of a dose-response curve visualizing the functional relationship between Akt and EGF. Users can generate a number
of graphs that are saved and can later be retrieved from the table at the top of the page. Generated graphs can also be saved on the computer and
used directly in a manuscript.
Helikar et al. BMC Systems Biology 2012, 6:96
Page 11 of 14
http://www.biomedcentral.com/1752-0509/6/96
is done by specifying the appropriate extracellular sig-
nal and output species (i.e., any species of interest) from
drop-down menus available on the page. On the generated
graph, the selected external species is represented on the
x-axis whereas the output species is represented on the
y-axis. Furthermore, similar to the real time simulation
feature, mutations to any of the cellular species can easily
be specified which allows users to simulate gain/loss-of-
function in an intuitive fashion. In the current version
of the software, users can generate the dose-response
graphs for all species in the model by selecting the appro-
priate input-output species. While we are in the course
of adding additional means of visualizing the simulation
results, users can also download all generated (raw) simu-
lation data, which can subsequently be analyzed by users
according to their needs.
The Dynamical Analysis feature can be used not only
to generate new hypotheses, but also to test the correct-
ness of the model. Because the models are built using
local knowledge of the individual interactions, how do
we know that all of this local information adds up to a
system that represents what is seen in the laboratory?
Hence the correctness of the model needs to be tested
on global phenomena of the system. The above exam-
ple demonstrates how the model of signal transduction
in a fibroblast cell can be tested to ensure that species
associated with apoptosis and growth (such as Akt) appro-
priately respond to a growth signal (EGF). If, for example,
the dose-response curve for Akt and EGF suggested a
negative correlation, one would have to go back and inves-
tigate which of the local interaction data resulted in the
contradictory result.
Seed models
In addition to the signal transduction model of a fibroblast
cell created and previously published by our group [32],
as part of our most recent research efforts, we have con-
structed additional models of the budding yeast cell cycle
[39] and host cell infection by Influenza A, including the
viral replication cycle (manuscript submitted). We have
also re-created a model of ErbB signaling and regulation of
the G1/S transition in the cell cycle during breast cancer.
This model was initially created by the authors to study
trastuzumab resistance and predict possible drug targets
in breast cancer [40]. All of these models are now avail-
able and published in The Cell Collective, hence available
to the scientific community as seed models for further
contributions and/or simulations and analyses.
Collaboration and accessibility
As discussed in the Background section, collaboration
amongst laboratory scientists working in different areas
of complex biological processes and the accessibility to
modeling frameworks is key to new discoveries using the
systems approach. These two properties were strictly kept
in mind when designing the software, and provide the
main framework for The Cell Collective.
First, motivated by this framework was the use a wiki-
like format to keep track of the knowledge concern-
ing the dynamical properties of biological process. This
framework was also applied to the way users interact with
the actual computational models.
Perhaps the most important feature in the context
of accessibility is the concept of “Published Models”
(Figure 8). These models created by the community are
freely accessible to all registered users, fostering the idea
of open science. All users can view the bio-logic as well
as the information in the knowledge base, and perform
real time simulations on these models directly. To make
changes to these models and see how these modifica-
tions affect the dynamics of the model, users can create
personal copies of published models. Once a copy of a
published model is created, the copy will be available
and visible only to the one user until shared under “My
Models” as seen in Figure 8. (As mentioned earlier, a
number of models are now available under Published
Models for all users to access and simulate.)
My Models is a collection of models created by any
given user. Users have an additional ability to share and
collaborate on any of these models with a select group
of colleagues. The degree to which such a collabora-
tion can take place is guided with the choice of three
types of permission a user can specify when sharing
his/her model. First, models can be shared such that
other users can simulate the shared models and view
the model’s bio-logic. A second way of model shar-
ing also allows other users to contribute to the mod-
els and directly edit them. Finally, models can be also
shared so that other users become model administrators
and have the same rights as the creator of the model,
including the ability to share the model with additional
collaborators.
Many biomedical research software tools (especially the
commercial ones) tend to limit users in such a way that
once the user commits to the tool, it becomes difficult
to move their data to a different platform. This is exactly
the opposite with The Cell Collective. In addition to being
able to share models with any and every user of the plat-
form, features to export models in formats that can work
with other modeling tools are also available. In the most
recent version, users can export all mathematical expres-
sions for each model (including the available published
models) in the form of flat text files as well as SBML
(SBML [28]).
Finally, a forum is available as part of The Cell Collective
modeling suite. This will afford users additional means of
communication with the scientific community as well as
with the platform’s development team.
Helikar et al. BMC Systems Biology 2012, 6:96
Page 12 of 14
http://www.biomedcentral.com/1752-0509/6/96
Figure 8 Main model panel. The filter in the top left corner allows the user to switch between the different types of models (discussed in text). The
majority of the space in the right section of the panel is dedicated to the model’s controls (boxed in) and more general information about the
model (e.g., creator and description). Users can also navigate from this panel to the simulation page as well as a page containing all model
constituents by using the Simulate and Model Bio-Logic buttons, respectively. As indicated in the right upper corner, users can also initiate the
creation of new models from this page.
Conclusions
Because of the inherent size and complexity of biochem-
ical networks, it is extremely difficult for a single person
or group to efficiently transfer the vast amount of labo-
ratory data into a mathematical representation; this fact
applies to any modeling technique. One way to address
this issue is to engage the community of laboratory sci-
entists that have generated these data and, hence, have
first-hand knowledge of the local protein-protein regula-
tory mechanisms. If the community of laboratory scien-
tists had a mechanism by which they could collaborate
and contribute their intimate knowledge of local inter-
actions into a large-scale global model, the creation of
these models would be greatly enhanced in terms of both
size and accuracy. As most laboratory scientists commu-
nicate their data in qualitative terms, rule-based models
which utilize such qualitative information provide an ideal
candidate for that platform.
Although qualitative models do not require an under-
standing of high level mathematics, it does assume that
users dealing with these models are familiar with rule-
based (e.g., Boolean) formalisms. At first, this may seem
a subtle issue (as most qualitative information generated
in laboratories is practically generated and interpreted in
Boolean terms; e.g., protein x AND y activate protein z),
however, the Boolean truth tables (and expressions) get
more complex as the size of the model increases. This
complexity effectively creates another challenge in build-
ing large-scale models. The Cell Collective and its major
component, Bio-Logic Builder (manuscript submitted),
aims at bridging this gap by enabling users to create these
dynamical models without having to directly interact with
the model’s mathematical complexities.
The collaborative nature of The Cell Collective also
opens doors to more open and reproducible science.
By integrating biological knowledge, currently dispersed
Helikar et al. BMC Systems Biology 2012, 6:96
Page 13 of 14
http://www.biomedcentral.com/1752-0509/6/96
across hundreds of scientific papers, scientists will be able
to test the integrity of this knowledge in the context of
the b/iological processes as a whole. The model building
process will make it easier to identify published results
that contradict each other, as well as find gaps in cur-
rent knowledge that may have not been realized. Using
a modeling platform such as The Cell Collective has the
potential to generate new hypotheses that can be further
verified in the laboratory.
Furthermore, the non-technical and easy-to-use nature
of building and simulating computational models in The
Cell Collective, the platform has a potential as a great edu-
cational tool for undergraduate and graduate biology stu-
dents with diverse mathematical/computer science skills.
Rather than studying biochemical pathways presented in
current textbooks as “static” and isolated components of
the cell, students can easily visualize and start understand-
ing cells as complex, dynamical systems – precisely as is
the case with real cells. Large models available in The
Cell Collective allow for the instruction of experimen-
tal design – because modeled biological processes have
(the complex) properties of the real counterparts, students
can learn how to design experimental studies, including
the concepts of controls. Students can also create simple
cellular models and study the dynamical properties of a
wide range of molecular subsystems such as positive and
negative feedback loops.
We are actively developing new features and making
The Cell Collective even more intuitive for users to inter-
act with it. We are also working on implementing a plug-in
system to allow the community to be directly involved in
the development of additional features.
Availability and requirements
The Cell Collective is platform independent, and can be
accessed through any modern web browser (Firefox and
Chromium are recommended). Data made public in The
Cell Collective are governed with GNU GPL v.3. The
platform is free for academic use.
Additional file
Additional file 1: Real time simulation example. Video example of a
real time simulation of a large-scale model of intracellular signal
transduction.
Competing interests
The authors declare that they have no competing interests.
Authors contributions
TH and JAR conceived the platform. TH designed the software and led the
development. BK, MS, SM, and KL developed the software. TH, JAR, KB, MS, SM,
KL, and AM tested the software. TH and JAR wrote the manuscript. All authors
read and approved the final manuscript.
Acknowledgements
This project was supported and funded by the College of Arts and Sciences at
the University of Nebraska at Omaha, the University of Nebraska Foundation,
and Patrick J. Kerrigan and Donald F. Dillon Foundations.
Author details
1Department of Mathematics, University of Nebraska at Omaha, Omaha, NE,
USA. 2College of Information Science and Technology, University of Nebraska
at Omaha, Omaha, NE, USA. 3Department of Pathology and Microbiology,
University of Nebraska Medical Center, Omaha, NE, USA.
Received: 27 March 2012 Accepted: 16 July 2012
Published: 7 August 2012
References
1.
Coveney PV, Fowler PW: Modelling biological complexity: a physical
scientist’s perspective. J R Soc Interface 2005, 2(4):267–80. [http://view.
ncbi.nlm.nih.gov/pubmed/16849185].
2.
Eungdamrong NJ, Iyengar R: Modeling cell signaling networks. Biol Cell
2004, 96(5):355–62. [http://view.ncbi.nlm.nih.gov/pubmed/15207904].
3.
Fisher J, Henzinger TA: Executable cell biology. Nat Biotechnol 2007,
25(11):1239–49. [http://view.ncbi.nlm.nih.gov/pubmed/17989686].
4.
Ideker T: Systems biology 101–what you need to know. Nat Biotechnol
2004, 22(4):473–5. [http://view.ncbi.nlm.nih.gov/pubmed/15085805].
5.
Kitano H: Systems biology: a brief overview. Science 2002,
295(5560):1662–4. [http://view.ncbi.nlm.nih.gov/pubmed/11872829].
6.
Kitano H: Computational systems biology. Nature 2002,
420(6912):206–10. [http://view.ncbi.nlm.nih.gov/pubmed/12432404].
7.
Laursen L: Computational biology: Biological logic. Nature 2009,
462(7272):408–10. [http://view.ncbi.nlm.nih.gov/pubmed/19940895].
8.
Powell K: All systems go. J Cell Biol 2004, 165(3):299–303. [http://view.
ncbi.nlm.nih.gov/pubmed/15123734].
9.
AFCS: Alliance for Cellular Signaling., 2011. [http://www.afcs.org].
10. STKE: Signal Transduction Knowledge Environment., 2008. [http://
stke.sciencemag.org].
11. Apweiler R, Bairoch A, Wu CH, Barker WC, Boeckmann B, Ferro S, Gasteiger
E, Huang H, Lopez R, Magrane M, Martin MJ, Natale DA, O’Donovan C,
Redaschi N, Yeh: UniProt: the Universal Protein knowledgebase.
Nucleic Acids Res 2004, 32(Database issue):D115—9. [http://view.ncbi.nlm.
nih.gov/pubmed/14681372].
12. Pico AR, Kelder T, van Iersel, M P, Hanspers K, Conklin BR, Evelo C:
WikiPathways: pathway editing for the people. PLoS Biol 2008,
6(7):e184. [http://view.ncbi.nlm.nih.gov/pubmed/18651794].
13. Smoot ME, Ono K, Ruscheinski J, Wang PL, Ideker T: Cytoscape 2.8: new
features for data integration and network visualization.
Bioinformatics 2011, 27(3):431–2. [http://view.ncbi.nlm.nih.gov/pubmed/
21149340].
14. Faeder JR: Toward a comprehensive language for biological systems.
BMC biology 2011, 9:68. [http://www.pubmedcentral.nih.gov/
articlerender.fcgi?artid=3195790&tool=pmcentrez&
rendertype=abstract].
15. KEGG: KEGG: Kyoto Encyclopedia of Genes and Genomes., 2011.
[http://www.genome.jp/kegg/].
16. Magrane M, Consortium U: UniProt Knowledgebase: a hub of
integrated protein data. Database (Oxford) 2011, 2011:bar009. [http://
view.ncbi.nlm.nih.gov/pubmed/21447597].
17. Reactome: Reactome., 2011. [http://www.reactome.org].
18. Cerami EG, Gross BE, Demir E, Rodchenkov I, Babur O, Anwar N, Schultz N,
Bader GD, Sander C: Pathway Commons, a web resource for
biological pathway data. Nucleic Acids Res 2011, 39(Database
issue):D685—90. [http://view.ncbi.nlm.nih.gov/pubmed/21071392].
19. Naldi A, Berenguier D, Faur´e A, Lopez F, Thieffry D, Chaouiya C: Logical
modelling of regulatory networks with GINsim 2.3. Biosystems 2009,
97(2):134–9. [http://view.ncbi.nlm.nih.gov/pubmed/19426782].
20. Albert I, Thakar J, Li S, Zhang R, Albert R: Boolean network simulations
for life scientists. Source Code Biol Med 2008, 3:16. [http://view.ncbi.nlm.
nih.gov/pubmed/19014577].
21. Saez-Rodriguez J, Alexopoulos LG, Epperlein J, Samaga R, Lauffenburger
DA, Klamt S, Sorger PK: Discrete logic modelling as a means to link
protein signalling networks with functional analysis of mammalian
Helikar et al. BMC Systems Biology 2012, 6:96
Page 14 of 14
http://www.biomedcentral.com/1752-0509/6/96
signal transduction. Mol Syst Biol 2009, 5:331. [http://view.ncbi.nlm.nih.
gov/pubmed/19953085].
22. M¨ussel C, Hopfensitz M, Kestler HA: BoolNet–an R package for
generation, reconstruction and analysis of Boolean networks.
Bioinformatics 2010, 26(10):1378–80. [http://view.ncbi.nlm.nih.gov/
pubmed/20378558].
23. Lloyd CM, Lawson JR, Hunter PJ, Nielsen: The CellML Model Repository.
Bioinformatics 2008, 24(18):2122–3. [http://view.ncbi.nlm.nih.gov/
pubmed/18658182].
24. Snoep JL, Olivier BG: Java Web Simulation (JWS); a web based
database of kinetic models. Mol Biol Rep 2002, 29(1-2):259–63. [http://
view.ncbi.nlm.nih.gov/pubmed/12241068].
25. Krishnamurthy L, Nadeau J, Ozsoyoglu G, Ozsoyoglu M, Schaeffer G, Tasan
M, Xu W: Pathways database system: an integrated system for
biological pathways. Bioinformatics 2003, 19(8):930–7. [http://view.ncbi.
nlm.nih.gov/pubmed/12761054].
26. Coskun SA, Qi X, Cakmak A, Cheng E, Cicek AE, Yang L, Jadeja R, Dash RK,
Lai N, Ozsoyoglu G, Ozsoyoglu ZM: PathCase-SB: integrating data
sources and providing tools for systems biology research. BMC syst
biol 2012, 6:67. [http://www.ncbi.nlm.nih.gov/pubmed/22697505].
27. Finney A, Hucka M: Systems biology markup language: Level 2 and
beyond. Biochem Soc Trans 2003, 31(Pt 6):1472–3. [http://view.ncbi.nlm.
nih.gov/pubmed/14641091].
28. Hucka M, Finney A, Sauro HM, Bolouri H, Doyle JC, Kitano H, Arkin AP,
Bornstein BJ, Bray D, Cornish-Bowden A, Cuellar AA, Dronov S, Gilles ED,
Ginkel M, Gor V, Goryanin II, Hedley WJ, Hodgman TC, Hofmeyr JH, Hunter
PJ, Juty NS, Kasberger JL, Kremling A, Kummer U, Le Nov`ere, N, Loew LM,
Lucio D, Mendes P, Minch E, Mjolsness ED, Nakayama Y, Nelson MR,
Nielsen PF, Sakurada T, SchaffJC, Shapiro BE, Shimizu TS, Spence HD,
Stelling J, Takahashi K, Tomita M, Wagner J, Wang J: The systems biology
markup language (SBML): a medium for representation and
exchange of biochemical network models. Bioinformatics 2003,
19(4):524–31. [http://view.ncbi.nlm.nih.gov/pubmed/12611808].
29. Nickerson D, Buist M: Practical application of CellML 1.1: The
integration of new mechanisms into a human ventricular myocyte
model. Prog Biophys Mol Biol 2008, 98:38–51. [http://view.ncbi.nlm.nih.
gov/pubmed/18606438].
30. CoLoMoTo: Common Logical Modelling Toolbox., 2011. [http://
compbio.igc.gulbenkian.pt/nmd/node/30].
31. Hu Z, Hung JH, Wang Y, Chang YC, Huang CL, Huyck M, DeLisi C: VisANT
3.5: multi-scale network visualization, analysis and inference based
on the gene ontology. Nucleic Acids Res 2009, 37(Web Server
issue):W115—21. [http://view.ncbi.nlm.nih.gov/pubmed/19465394].
32. Helikar T, Konvalina J, Heidel J, Rogers J A: Emergent decision-making in
biological signal transduction networks. Proc Natl Acad Sci U S A 2008,
105(6):1913–8. [http://www.ncbi.nih.gov/pubmed/18250321].
33. Helikar T, Rogers JA: ChemChains: a platform for simulation and
analysis of biochemical networks aimed to laboratory scientists. BMC
Syst Biol 2009, 3:58. [http://view.ncbi.nlm.nih.gov/pubmed/19500393].
34. Barabasi A, Albert R: Emergence of scaling in random networks.
Science (New York, N.Y.) 1999, 286(5439):509–12. [http://www.ncbi.nlm.
nih.gov/pubmed/10521342].
35. Aldana M, Cluzel P: A natural class of robust networks. Proc Natl Acad
Sci U S A. 2003, 100(15):8710–4. [http://www.ncbi.nlm.nih.gov/pubmed/
12853565].
36. Szalay A, Gray J: 2020 computing: science in an exponential world.
Nature 2006, 440(7083):413–4. [http://view.ncbi.nlm.nih.gov/pubmed/
16554783].
37. Jiang T, Qiu Y: Interaction between Src and a C-terminal proline-rich
motif of Akt is required for Akt activation. J Biol Chem 2003,
278(18):15789–93. [http://view.ncbi.nlm.nih.gov/pubmed/12600984].
38. Howe A, Aplin AE, Alahari SK, Juliano RL: Integrin signaling and cell
growth control. Curr Opin Cell Biol 1998, 10(2):220–31. [http://view.ncbi.
nlm.nih.gov/pubmed/9561846].
39. Irons D: Logical analysis of the budding yeast cell cycle. Journal of
Theoretical Biology 2009, 257(4):543–59. [http://www.ncbi.nlm.nih.gov/
pubmed/19185585].
40. Sahin O, Fr¨ohlich H, L¨obke C, Korf U, Burmester S, Majety M, Mattern J,
Schupp I, Chaouiya C, Thieffry D, Poustka A, Wiemann S, Beissbarth T, Arlt
D: Modeling ERBB receptor-regulated G1/S transition to find novel
targets for de novo trastuzumab resistance. BMC Syst Biol 2009, 3:1.
[http://view.ncbi.nlm.nih.gov/pubmed/19118495].
doi:10.1186/1752-0509-6-96
Cite this article as: Helikar et al.: The Cell Collective: Toward an open and
collaborative approach to systems biology. BMC Systems Biology 2012 6:96.
Submit your next manuscript to BioMed Central
and take full advantage of:
• Convenient online submission
• Thorough peer review
• No space constraints or color figure charges
• Immediate publication on acceptance
• Inclusion in PubMed, CAS, Scopus and Google Scholar
• Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
|
22871178
|
STAT1 = ( Jak1 ) OR ( IL27 AND ( ( ( NFAT ) ) ) )
IL23R = ( ( IL23 AND ( ( ( STAT3 ) ) ) ) AND NOT ( Tbet ) ) OR ( STAT3 )
Foxp3 = ( ( ( ( TGFbR ) AND NOT ( GATA3 ) ) AND NOT ( IL6R AND ( ( ( STAT3 ) ) ) ) ) AND NOT ( IL21R ) ) OR ( ( ( ( STAT5 ) AND NOT ( GATA3 ) ) AND NOT ( IL6R AND ( ( ( STAT3 ) ) ) ) ) AND NOT ( IL21R ) )
IL4 = ( ( GATA3 AND ( ( ( NFAT ) ) ) ) AND NOT ( STAT1 ) )
IL12R = ( ( STAT4 ) AND NOT ( GATA3 ) ) OR ( ( TCR ) AND NOT ( GATA3 ) ) OR ( Tbet ) OR ( IL12 AND ( ( ( NFAT ) ) ) )
RORgt = ( ( ( ( TGFbR AND ( ( ( STAT3 AND IL6R ) ) OR ( ( IL21R AND STAT3 ) ) ) ) AND NOT ( GATA3 ) ) AND NOT ( Tbet ) ) AND NOT ( Foxp3 ) )
SOCS1 = ( Tbet ) OR ( STAT1 )
IL2 = ( ( NFAT AND ( ( ( NFkB ) ) ) ) AND NOT ( Tbet ) )
NFAT = ( ( TCR ) AND NOT ( Foxp3 ) )
IFNgR = ( IFNg AND ( ( ( NFAT ) ) ) ) OR ( IFNg_e AND ( ( ( NFAT ) ) ) )
Tbet = ( ( ( STAT4 ) AND NOT ( RORgt ) ) AND NOT ( Foxp3 ) ) OR ( ( ( STAT1 ) AND NOT ( RORgt ) ) AND NOT ( Foxp3 ) ) OR ( ( ( Tbet AND ( ( ( NOT IFNg AND NOT IL12 ) ) ) ) AND NOT ( RORgt ) ) AND NOT ( Foxp3 ) )
STAT3 = ( IL21R ) OR ( IL6R ) OR ( IL23R )
IL2R = ( IL2 AND ( ( ( NFAT ) ) ) )
GATA3 = ( ( ( ( ( STAT5 ) AND NOT ( Tbet ) ) AND NOT ( RORgt ) ) AND NOT ( TGFb ) ) AND NOT ( Foxp3 ) ) OR ( ( ( ( ( STAT6 AND ( ( ( NFAT ) ) ) ) AND NOT ( Tbet ) ) AND NOT ( RORgt ) ) AND NOT ( TGFb ) ) AND NOT ( Foxp3 ) ) OR ( ( GATA3 ) AND NOT ( Tbet ) )
IL6R = ( IL6 ) OR ( IL6_e )
IL17 = ( ( RORgt ) AND NOT ( STAT1 ) ) OR ( ( ( STAT3 AND ( ( ( IL17 ) ) AND ( ( IL23R ) ) ) ) AND NOT ( STAT5 ) ) AND NOT ( STAT1 ) )
TGFbR = ( TGFb AND ( ( ( NFAT ) ) ) )
IL4R = ( ( IL4 ) AND NOT ( SOCS1 ) ) OR ( IL4_e )
IRAK = ( IL18R )
IFNg = ( ( ( STAT4 AND ( ( ( NFkB ) ) AND ( ( NFAT ) ) ) ) AND NOT ( STAT6 ) ) AND NOT ( STAT3 ) ) OR ( ( Tbet ) AND NOT ( STAT3 ) ) OR ( NFkB )
IL21 = ( STAT3 AND ( ( ( NFAT ) ) ) )
STAT5 = ( IL2R )
NFkB = ( ( IRAK ) AND NOT ( Foxp3 ) )
IL18R = ( ( IL18 AND ( ( ( IL12 ) ) ) ) AND NOT ( STAT6 ) )
STAT4 = ( ( IL12R AND ( ( ( IL12 ) ) ) ) AND NOT ( GATA3 ) )
STAT6 = ( ( ( IL4R ) AND NOT ( SOCS1 ) ) AND NOT ( IFNg ) )
IL21R = ( IL21 )
IL6 = ( RORgt )
Jak1 = ( ( IFNgR ) AND NOT ( SOCS1 ) )
|
BIOINFORMATICS
Vol. 28 ECCB 2012, pages i495–i501
doi:10.1093/bioinformatics/bts410
Boolean approach to signalling pathway modelling in
HGF-induced keratinocyte migration
Amit Singh1,2,†, Juliana M. Nascimento1,2,†, Silke Kowar1,2, Hauke Busch1,2,‡,∗
and Melanie Boerries1,2,‡,∗
1Freiburg Institute for Advanced Studies, LifeNet, Albert-Ludwigs-University of Freiburg, Albertstrasse 19 and
2Center for Biological Systems Analysis, Albert-Ludwigs-University of Freiburg, Habsburger Strasse 49, 79104
Freiburg, Germany
ABSTRACT
Motivation: Cell migration is a complex process that is controlled
through the time-sequential feedback regulation of protein signalling
and
gene
regulation.
Based
on
prior
knowledge
and
own
experimental data, we developed a large-scale dynamic network
describing the onset and maintenance of hepatocyte growth factor-
induced migration of primary human keratinocytes. We applied
Boolean logic to capture the qualitative behaviour as well as short-
and long-term dynamics of the complex signalling network involved
in this process, comprising protein signalling, gene regulation and
autocrine feedback.
Results: A Boolean model has been compiled from time-resolved
transcriptome data and literature mining, incorporating the main
pathways involved in migration from initial stimulation to phenotype
progress.
Steady-state
analysis
under
different
inhibition
and
stimulation conditions of known key molecules reproduces existing
data and predicts novel interactions based on our own experiments.
Model simulations highlight for the first time the necessity of a
temporal sequence of initial, transient MET receptor (met proto-
oncogene, hepatocyte growth factor receptor) and subsequent,
continuous epidermal growth factor/integrin signalling to trigger and
sustain migration by autocrine signalling that is integrated through
the Focal adhesion kinase protein. We predicted in silico and verified
in vitro that long-term cell migration is stopped if any of the two
feedback loops are inhibited.
Availability: The network file for analysis with the R BoolNet library
is available in the Supplementary Information.
Contact: melanie.boerries@frias.uni-freiburg.de
or hauke.busch@frias.uni-freiburg.de
Supplementary information: Supplementary data are available at
Bioinformatics online.
1
INTRODUCTION
Cell
migration
and
wound
healing
are
complex
cellular
processes that involve keratinocytes, fibroblasts, blood vessels and
inflammatory cells (Xue et al., 2007). Keratinocyte migration
plays an important role in re-epithelialization and wound healing
(Hunt et al., 2000), which is a multistep cellular process by the
†The authors wish it to be known that, in their opinion, the first two authors
should be regarded as joint First Authors.
‡The authors wish it to be known that, in their opinion, the last two authors
should be regarded as joint Last Authors.
∗To whom correspondence should be addressed.
coordination of extra- and intracellular signals (Muyderman et al.,
2001; Werner et al., 2007). The precise regulation of cell migration
in its temporal sequence, activation and de-activation is crucial for
tissue homeostasis. In its aberrant form, it can lead to scar formation
(Heng, 2011) and has critical implications to cancer metastasis
formation (Schäfer and Werner, 2008).
Different growth factors such as hepatocyte growth factor (HGF),
epidermal growth factor (EGF), transforming growth factor-beta
(TGF-β), keratinocyte growth factor (KGF) and fibroblast growth
factor (FGF) that activate and regulate cell migration have been
extensively studied in many cell types (Birchmeier et al., 2003;
Hudson and McCawley, 1998; Jaakkola et al., 1998; Pastore et al.,
2008; Tsuboi et al., 1993). These growth factors have been found to
overlap with mitogen-activated protein kinase (MAPK) pathways
(Cho and Klemke, 2000; Kain and Klemke, 2001; Klemke et al.,
1997).
HGF interacts and activates MET receptor (Bottaro et al.,
1991) to induce context-dependent several cellular processes such
as proliferation, cell movement or morphogenic differentiation
(Brinkmann et al., 1995; Clague, 2011; Jeffers et al., 1996; Medico
et al., 1996). Herein, we focus on HGF-induced migration of primary
normal human keratinocytes (NHK).
Although there is vast literature concerning HGF-induced
keratinocyte migration and MET receptor dynamics, the dynamic
interplay of initial MET receptor regulation and subsequent
autocrine regulation that initiate, sustain and control cell migration
remain poorly understood. Based on time-resolved transcriptome
data of NHK after HGF stimulation, we have previously inferred a
gene regulatory model describing the decision process of NHK cells
towards migration (Busch et al., 2008). From the model analysis it
was evident that several pathways coordinate their action to initiate
and sustain cell migration upon initial HGF stimulation: migration is
started through the AP-1 system and maintained after MET receptor
internalization (Clague, 2011) by autocrine signalling through EGF
receptor (EGFR) and urokinase plasminogen activator surface
receptor (uPAR) (Schnickmann et al., 2009). The model predicted
qualitatively how the temporal sequence of transient MET receptor
activation and subsequent long-term EGF receptor activity sustained
the migratory phenotype. However, as the model was based on
transcriptome data alone, there was no mechanistic explanation
of the observed processes. A model combining transcriptome data
with mechanistic protein signalling has been missing so far. A
major obstacle in building such a model lies in the different time
scales involved in the process of cell migration. In general, the
transcriptome response changes over several hours, while protein
signalling pathways become active within minutes upon receptor
© The Author(s) 2012. Published by Oxford University Press.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which
permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
A.Singh et al.
HGF
EGFR
DAG
PKC
AKAP12
cMYC
EGR1
CRKL
IP3
Ca2+
RSK
CREB
ATF2
CCL20
HBEGF
CTGF
IL8
DOCK1
C3G
RAP1
FAK
Integrins
MMP1/10
ECM
Plasmin
uPA
AKT
CDC42/
RAC1
MEKK1
MEKK7
JNK
PAK2
PAK3
PAK1
MEKK4
MKK4
MLK3
MKK3
MKK6
p38
cFOS
PTGS2
cJUN
Cell Migration
ELK1
ETS
STAT3
CyclinD
CDK2
CDKN2A
CDKN1A
Proliferation
uPAR
PAI-1
PLC
PI3K
PTEN
MET
SHC
GRB2
RAS
RAF
MEK
ERK
SOS
AP1
DUSP1
PTGS2
cJUN
Gene
Protein
Drain
Input
Activation
Inhibition
"AND" Gate
PLC/PKC
MAPK
uPA/uPAR
AP-1
System
PI3K
Jnk/p38
Rac1/CDC42
Fig. 1. Boolean network model of the HGF-induced keratinocyte migration. Nodes are connected by directed edges, where black and red connections denote
activating and inhibitory interactions, respectively. Red nodes represent transcriptionally regulated proteins, yellow nodes are endpoints of the network. ‘AND’
‘gates are denoted by blue dots and ‘OR’ gates are found where more than one edge connects to single node. Dashed edges denote interactions that have not
been considered, when calculating the steady state and are shown for completeness
stimulation (Mesecke et al., 2011). Capturing all necessary and
sufficient events on the protein signalling level, including kinetic
parameters, is close to impossible by current biological technology.
To link our prior transcriptome-based model with protein
signalling pathways, we present a Boolean network model of HGF-
induced keratinocyte migration. The Boolean approach allows to
derive important functional properties and predictions without the
need for detailed quantitative kinetic data and parameters. In the
past, the approach has been successfully applied for diverse systems
such as gene regulatory networks (Albert and Othmer, 2003; Chaves
et al., 2005), models of floral morphogenesis (Mendoza et al., 1999),
mammalian cell cycle (Mendoza, 2006), EGFR signalling (Samaga
et al., 2009) or apoptosis (Schlatter et al., 2009).
To our knowledge, this is the first model for HGF-induced
keratinocyte migration that incorporates protein signalling, gene
regulation and autocrine feedback, following cellular dynamics
from initial stimulation to the execution of the phenotype. To
obtain the dynamical behaviour reproducing literature knowledge
and our own experimental data, we include several time scales in
the model mimicking the fast activation of downstream signalling
of MET, MAPK/ERK and p38/JNK pathways, as well as the
slow transcriptome response and subsequent autocrine activation
of EGFR and uPA receptors, all of which are necessary to sustain
cell migration after MET receptor internalization. Specifically, from
a logical steady-state analysis, we show that priming of the HGF–
MET receptor system is necessary for continued autocrine regulation
i496
Boolean modelling of keratinocyte migration
through EGFR and integrins, sustaining the MAPK/ERK activity.
More importantly, we predicted and showed experimentally, that the
inhibition of the plasminogen activator inhibitor-1 (PAI-1), or serpin
E1, a serine protease inhibitor, stops cell migration only beyond 1 h
of stimulation, when autocrine signalling loops through uPA/uPAR
become important and after the first wave of protein signalling and
transcriptional response.
2
METHODS
2.1
Reconstruction of the NHK migration model
A Boolean network, comprising protein signalling pathway, gene expression
dynamics and autocrine feedback was constructed based on our previous
gene regulatory model for keratinocyte migration (Busch et al., 2008).
There, time-resolved gene expression data of NHK were recorded at t =
[0h,1h,2h,3h,4h,6h,8h] after stimulation with HGF (ArrayExpress ID:
E-TABM-440). As a basis for our HGF-induced cell migration network,
we chose genes that have either a large differential response after HGF
stimulation or genes that are known to be functionally related to cell
migration. Genes finally included in the model are depicted in Figure
2A. To link immediate early and late responding genes to initial MET
receptor signalling and subsequent protein pathways, respectively, we
integrated differentially expressed genes with signalling pathways known
from literature. Additional pathways for the HGF migration network
were identified through the commercial IPA (Ingenuity Systems; www.
ingenuity.com) software. A dataset containing gene identifiers and
corresponding expression values was uploaded into the application. Each
gene kinetic was mapped to its corresponding object in the Ingenuity
Knowledge Base. The above identified molecules were overlaid onto a global
molecular network developed from information contained in the application.
This generated a score with meaningful and significant networks, biological
functions and the canonical pathways based on the Fisher exact test.
2.2
The NHK migration model as a dynamic
Boolean model
We used a Boolean model framework to construct a dynamic, temporally
discrete model for NHK migration. Each node y can take the values 0 or 1,
representing either the present/absent or inactive/active, i.e. phosphorylated,
state of the protein or gene. The network state is represented by the vector
with the set of Boolean variables Y ={y1,y2,...,yn}, where yi denotes the
state of the ith node. The state of activation of each node changes according
to the transition function F ={f1,f2,...,fn}. The next state of the network
Y(t+1) changes in discrete time steps according to yi(t+1)=fi{x(t)}.
We simulated the Boolean network under synchronous update using the R
BoolNet library (Müssel et al., 2010). As we consider both protein signalling
and gene regulation, two time scales were included in the model. Rapid
protein modifications such as phosphorylation can thereby be separated from
long-term effects, transcriptional changes, protein synthesis and autocrine
signalling. Time-scale separation was done by first introducing a reference
time through the transcriptome kinetics and equating the time of maximal fold
change of the respective genes with their switching-on time. Consequently,
our model contains two time scales: 0–1 and 1–3 h (marked as 1 and 3,
respectively), denoting the time intervals after HGF stimulation, during
which the reactions can be switched on the earliest. These represent the
early HGF downstream signalling and first transcriptional response as well
as the autocrine feedback, which are both necessary to trigger and sustain
cell migration.
The reference publications from which the interactions have been inferred
as well as their Boolean transition functions and time windows are listed in
the Supplementary Table S1.
2.3
Cell culture
Normal human skin keratinocytes (NHK) were derived from foreskin
epidermis and cultivated in keratinocyte serum free medium (KSFM;
Invitrogen, Carlsbad, CA, USA) as previously described (Busch et al., 2008).
Cells were kept under a humidified environment with 5% CO2 and 37◦C.
NHK up to Passages 4–5 were used in all experiments of this study. Cells
were treated as described bellow and collected after 1, 2, 3, 4, 6 and 8 h
treatment for expression profiling.
2.4
Scratch assay
Monolayer scratch assays were used to evaluate migration of NHK
as described before (Busch et al., 2008). Briefly, cells were grown
to confluence in ibidi μ-dish containing culture inserts (ibidi, Munich,
Germany) and treated with mitomycin c 10 μg/mL (Sigma-Aldrich, St.
Louis, MO, USA) for 3 h before stimulation. Cells were stimulated with
10 ng/mL HGF (Sigma-Aldrich) and/or 25 μM Tiplaxtinin, a PAI-1 inhibitor
(Axon Medchem, Groningen, Netherlands). Time-lapse microscopy of cell
migration was recorded using the Perfect Focus Systems , every 30 min up to
24 h, on a Nikon Eclipse Ti microscope with a Digital Sight DS-QiMc (Nikon
Instruments Inc., Tokyo, Japan), coupled to an ibidi-heating chamber.
2.5
Cell migration assay
Keratinocyte migration was analysed with an independent second technique,
so-called xCELLigence Real-Time Cell Analyzer DP (Roche Diagnostics).
The specific migration CIM-plates were filled with medium and the
respective stimulus/inhibitor as described above. NHK were seeded (6×104
cells/well) into the top chamber wells of the CIM-plate according to the
manufacturer instructions. Cell migration was monitored every 15 min for
up to 24 h by changes of the impedance signal of the cells that crossed the
membrane from the top to the bottom chamber. For analysis of migration, the
area under the curve was measured for the first 8 h. The data were expressed
as the mean ± SD of quadruplicates in three independent experiments.
Differences were assessed by the Student t-test for unpaired samples and
a P-value <0.05 was considered to be significant.
2.6
Western blot
NHK were lysed in RIPA buffer containing protease inhibitor cocktail
(Roche), and later diluted in Laemmli buffer. Proteins were electrophoresed
on 12.5% sodium dodecyl sulphate (SDS)-polyacrylamide gels, transferred
to polyvinylidene difluoride membranes, and immunoblotted with antibodies
to total p44/42 MAPK (ERK1/2) (Cell Signalling #9102), phospho p44/42
MAPK (pERK1/2) (Cell Signalling #9101), total FAK (Cell Signalling
#3285), phospho-FAK (Tyr925) (Cell Signalling #3284) overnight at
4◦C. Membranes were visualized with chemiluminescence after using
Table
1. Predicted
network
steady
states
under
different
network
perturbations
HGF EGF Inhibition Over-expression Cell migration
Confirmed
1
0
—
—
1
Own data
0
1
—
—
0
Predicted
0
1
FAK
—
0
Predicted
1
1
EGFR
—
0
Own data
1
0
uPAR
—
0
19020551
1
0
PAI-1
—
0
Own data
1
0
PTGS2
—
0
Own data
1
0
IL8
—
0
Own data
1
0
—
AKAP12
0
21779438, Own data
1
0
—
PTEN
0
16246156
The last column lists the PubMed IDs of the respective publication.
i497
A.Singh et al.
0h
1h
2h
3h
4h
6h
8h
egr1
0h
1h
2h
3h
4h
6h
8h
il8
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
fos
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
ptgs2
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
pak3
0h
1h
2h
3h
4h
6h
8h
hbegf
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
dusp1
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
ctgf
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
ccl20
0h
1h
2h
3h
4h
6h
8h
akap12
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
mmp10
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
mmp1
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
itga2
0h
1h
2h
3h
4h
6h
8h
serpine1/PAI-1
0
1
2
3
4
0h
1h
2h
3h
4h
6h
8h
plaur/uPAR
0
1
2
3
4
HGF
MET
SHC
GRB2
SOS
RAS
RAF
MEK
ERK
PLCG
IP3
DAG
CA
PKC
CRKL
DOCK1
PI3K
AKT
RAP1
C3G
PAK1
PAK2
PAK3
CDC42RAC1
MLK3
RSK
CREB
CMYC
EGR1
ELK1
ETS
CDK2
CDKN1A
CDKN2A
CYCLIND
PROLIFERATION
STAT3
CFOS
CJUN
AP1
CCL20
COX2
IL8
CTGF
ATF2
HBEGF
EGFR
MKK3
MKK4
MKK6
P38
JNK
MEKK7
MEKK1
MEKK4
UPAR
UPA
PAI1
PLASMIN
MMP
ECM
INTEGRIN
FAK
CELL_MIGRATION
4
Time
0h
1h
3h
a
b
c
d
MAPK/ERK
PLC/PKC
PI3K
CDC42/RAC1
Proliferation
AP-1
Transcriptome
Response
e
JNK/p38
uPA/uPAR
HGF/MET
egr1
il8
fos
ptgs2
ctgf
dusp1
hbegf
pak3
ccl20
akap12
mmp10
mmp1
plaur/uPAR
serpine1/
PAI-1
itga2
Time [h]
Fold Change [log2]
0
1
2
3
4
0
1
2
3
4
0
1
2
3
4
0
1
2
3
4
s.s.
EGFR Inhibition
PAI-1 Inhibition
A
B
Fig. 2. (A) Transcriptome time series of differential regulation with respect to 0 h time point for genes included in the Boolean cell migration network.
(B) Network simulation of time sequential pathway activation. (a) Path to attractor upon HGF stimulation up to 1 h after stimulation. (b) Change in network
state after autocrine signalling through uPA and integrin signalling up to 3 h after stimulation, initializing the network using the network state after 1 h and
setting PAI-1 activity to 1. (c) Switching off MET receptor signalling after 3 h. Cell migration sustains in steady state (s.s.). (d) and (e) Inhibition of EGFR and
PAI-1, respectively, in both cases leading to a stop in cell migration. Time increases in arbitrary units from left to right until a logical steady state is reached.
Absolute times correspond to the maximal fold induction of the corresponding transcriptome data. Pathway-based grouping of network nodes (cf. Fig. 1) is
indicated in colour and named on the left
the appropriate horseradish peroxidase-linked secondary antibody (Sigma-
Aldrich). Immunoblots were quantified using Multi Gauge v3.1 (Fujifilm)
software. Values obtained for both p44 and p42 ERK bands were added
together and phospho p44/42 values were normalized to total p44/42. Values
of phospho-FAK (Tyr925) were normalized to total FAK.
3
RESULTS AND DISCUSSION
Upon
stimulation
with
HGF
keratinocytes
start
to
migrate
collectively.
Several
points
of
interference
that
modulate
keratinocyte migration have been previously identified. However,
time sequential orchestration of the whole-cell signalling remains
unclear so far. Our goal is to understand how downstream signalling
of the HGF-activated MET receptor is translated into a sustained
migratory behaviour. To accomplish this, we developed a Boolean
network model of the combined MAPK signalling pathways, gene
regulation and autocrine feedback, which links known interactions
of downstream protein and gene targets of MET with subsequent
changes in the cellular homeostasis.
3.1
Boolean network model properties
The keratinocyte migration model is a logical interaction hypergraph
connected by logic gates. It comprises 66 nodes, excluding the drain
nodes and 66 interactions (Fig. 1), integrating the main pathways and
genes known to be involved in HGF-induced keratinocyte migration.
For detailed information about the biological processes and context
from which the model was derived, please confer to Supplementary
Table S1.
There are two input nodes, the MET receptor, stimulated through
HGF, as well as EGFR, stimulated by HBEGF. Both receptors
have been shown to be involved in keratinocyte migration in a
time-sequential manner (Busch et al., 2008). Four further nodes
are included to applying external interventions: PAI-1, AKAP12,
DUSP1 and PTEN. AKAP12 and DUSP1 are not included in
the calculation of the steady-state below. Despite the fact that
they are found to be up-regulated on the transcriptome level, the
respective proteins seem to exert a stabilizing negative feedback
function (Legewie et al., 2008), not completely inhibiting their
specific targets, but allowing for a rapid protein induction (Blüthgen,
2010). In line with this, we find akap12 strongly up-regulated
in HGF-induced keratinocyte migration (Fig. 2A), although over-
expression of akap12 has been associated with reduced motility
(Gelman, 2010).ABoolean representation of such negative feedback
behaviour is not straight forward and will often lead to — a
biologically questionable — oscillatory steady state. Hence, we
included the proteins for completeness, yet excluded it from steady-
state analysis.
3.2
Dynamics of the Boolean network model for
migration
To search for steady-state attractors of the Boolean network model,
we randomly initialized all network nodes, except for HGF/PAI-1
and AKAP12/PTEN with either 0 or 1 and performed synchronous
state transitions until a simple attractor was reached. Activating the
nodes HGF and PAI-1 to 1 mimicked HGF stimulation and allowed
for autocrine feedback through uPAR signalling. Setting AKAP12
and PTEN to zero excluded their inhibitory effects. Sampling
over n=107 random initialization of the network we find only
one attractor of the network with the node CELL MIGRATION
switched on. Although only a minor fraction of all possible
i498
Boolean modelling of keratinocyte migration
0h
1h
3h
8h
HGF
HGF/PAI-1
inhibitor
Control
HGF
HGF + Inhibitor
0
1
2
3
***
***
AUC (8h)
Control
HGF
HGF + Inhibitor
0
2
4
6
8
0.8
1.0
1.2
1.4
1.6
1.8
Time (h)
Delta Cell Index
ERK
p-ERK
C
H
H+I
C
H
H+I
C
H
H+I
C
H
H+I
1h
2h
3h
8h
C
H
H+I
C
H
H+I
C
H
H+I
C
H
H+I
0.0
0.5
1.0
1.5
2.0
1h
2h
3h
8h
*
*
*
*
pERK/ERK
A
B
C
Fig. 3. Keratinocyte migration analysis under HGF and PAI-1 inhibition. (A) Life-time imaging microscopy of NHK up to 8 h. HGF stimulation increases
migration of NHK (upper panel), addition of PAI-1 inhibitor, Tiplaxtinin, decreases strongly the HGF effect (lower panel). Bar: 20 μm. (B) Increased HGF
migration is significantly reduced with PAI-1 inhibitor (25 μM) addition after a short time period (left plot). Migration response is calculated as area under
the curve (AUC; right plot). (C) Determination of pERK level under HGF and inhibition conditions. Original western blot of pERK and ERK for different
time points reveals a continuous activation of pERK under HGF (H) stimulation over time comparing to control (C). In contrast under PA1-1 inhibition (H+I)
the pERK level decrease significantly over time. ∗denote a t-test P-value <0.05, data points obtained in triplicate. The ratio of pERK/ERK is shown for the
respective time points (right plot)
network initializations (n=262) has been sampled, this result is still
suggestive of a robust response irrespective of the initial network
state, i.e. cell migration follows upon MET receptor activation and
subsequent autocrine signalling.
In detail node and pathway activity moves along the following
steps after transient MET receptor signalling: (i) the first and
necessary input into the network towards migration is the
stimulation with HGF, which immediately and specifically activates
the MET receptor. Within the first hour, the signal activates
three different downstream pathways, PLC/PKC, MAPK/ERK
and PI3K (grey boxes, Fig. 1); (ii) ERK phosphorylation
activates transcriptional responses, leading to the down-regulation
of proliferation and activates, together with p38/JNK, essential
cytokines and transcription factors for migration within the first
hour, such as HBEGF, IL8 and ATF2, as well as cJUN and
cFOS, respectively. HBEGF has been shown to mediate the
subsequent autocrine activation of EGFR (Busch et al., 2008); (iii)
The activation of cJUN and cFOS nodes leads to an initiation
of the AP-1 system, which in turn stimulates the uPA/uPAR
signalling pathway by activation of the uPAR, triggering the
formation from plasminogen activator to plasmin. According to
the transcriptome kinetics, the uPA/uPAR pathway becomes active
2 h after HGF stimulation, being controlled by PAI-1. Plasmin
is a major factor for induction of metalloproteinases 1 and 10
(MMP1/MMP10), linking degradation of the extracellular matrix
(ECM) with integrin signalling; (iv) The integrins transmit the
extracellular signalling back into the cell through the focal adhesion
kinase (FAK). Together with activated EGFR this protein sustains
PLC/PKC, MAPK and PI3K activity similar to the initial HGF/MET
activation. It is known that the MET receptor undergoes rapid
internalization, possibly switching off its signalling in favour of
EGFR activity. In fact, we have shown previously that HGF/MET
i499
A.Singh et al.
activity is not required for keratinocyte migration beyond 1.5 h after
stimulation (Busch et al., 2008). Accordingly, the model suggests
that continued PI3K up-regulation can only occur through the
combined activity of integrin and EGFR signalling in the presence
of FAK. Signalling continues through AKT, DOCK1 and RAS to
sustain RAC1/CDC42 activity, which result in the downstream
activity of p21 protein (CDC42/RAC)-activated kinases (PAKs),
mitogen-activated kinase kinases (MKK) and finally activation of
JNK/p38. This closes the autocrine loop through time sequentially
activated external receptors. This late response of the keratinocyte
migration network through activation of uPAR, integrins and EGFR
triggers similar pathways as the MET signalling, but furthermore
results in the prolonged activation of PLC/PKC, MAPK/ERK, PI3K,
RAC1/CDC42 and JNK/p38 pathways to sustain the long-term
migration response.
Figure 2B shows a simulation of the network dynamics after
HGF stimulation up to 3 h, as marked by the white vertical
gaps, having initialized all network nodes, except for HGF with
0 for clarity. The transitions towards the final steady states are
indicated in arbitrary time units. Clearly, an activation wave of
the three downstream pathways PLC/PKC, MAPK/ERK and PI3K
is evident, resulting in the up-regulation of the respective target
genes (Fig. 2A), the AP-1 system and starting cell migration within
1 h. In the following 2 h, autocrine signalling loops activate EGFR
and integrin signalling through HBEGF and uPAR, respectively.
Switching off the MET receptor has no influence on the steady
state at this time, and cell migration continues. Switching off
either uPA/uPAR signalling through PAI-1 or inhibiting EGFR stops
migration through subsequent switching off all pathways.
Table 1 lists the steady states of the Boolean network under
different input settings and/or different scenarios. Interestingly, the
model predicts that cells will migrate only when HGF signalling
primes the cells, followed by EGFR and integrin signalling. This is
in line with our previous findings, which now can be explained on
the causal level of protein signalling.
Lastly, we note that our model still has limitations predicting
the correct long-term behaviour for transiently activated genes such
as egr1, il8, ctgf or ccl20. Although the model captures the long-
term cell migration response and upstream pathway activity, it does
not yet include a negative feedback down-regulating of these genes
(compare Fig. 2A and B). Although the biological consequences of
the transient gene activation remain unclear so far, this divergence
between the model steady-state behaviour and experimental data
beyond 3 h need to be addressed in more detail in the future.
Model simulation clearly reflects the necessity for the time
sequential pathway activation, shown by the early and late
steady state after 1 and 3 h, respectively. Initial HGF/MET
receptor signalling triggers, while subsequent integrin/EGF receptor
signalling sustains MAPK/ERK, PI3K, PLC/PKC and JNK/p38
signalling (Fig. 3C). Model simulations further predict that
continued migration depends on both EGFR and integrin signalling.
Failure of either one causes the down-regulation of the FAK protein
and subsequent PI3K, MAPK/ERK and p38/JNK pathways (Fig.
2, Columns d and e). Indeed, we have previously shown the
dependency of cell migration on sustained EGFR activity after MET
signalling pathway activation (Busch et al., 2008).
To experimentally validate the long-term dependency of NHK
on the FAK protein-mediated integrin signalling, we interrupt the
uPA/uPAR signalling pathway through inhibition of PAI-1. In line
with previous findings (Providence and Higgins, 2004) and predicted
from our network simulation, long-term, but not immediate cell
migration should be decreased. A scratch assay (Fig. 3A) shows
a significant decrease with the usage of the PAI-1 inhibitor
(Tiplaxtinin) in migration when compared with control conditions.
While under HGF stimulation, the scratch is almost closed after
8 h, hardly any cell movement is detected under additional PAI-1
inhibitor treatment. The delayed impact of PAI-1 inhibition becomes
evident from a real-time analysis of keratinocyte migration using the
xCELLigence Cell Analyzer (Fig. 3B). Clearly, the migration speed
becomes strongly reduced after 2.5 h, in line with the suggested
role for ‘late’uPA/uPAR signalling pathway involvement, activating
FAK through integrin/EGFR signalling for sustained migration. This
time point also coincides with the start of transcriptional activation of
PAI-1 (Fig. 2A), furthermore supporting the late involvement of this
pathway. Interestingly, MET receptor and integrin/FAK pathways
both activate MAPK/ERK, PI3K and PLC/PKC, RAC1/CDC42
and JNK/p38. However, while the former primes the cells towards
migration, the impact of the latter on downstream pathways seems
to be much more pronounced. Analysing the phosphorylation state
of ERK (pERK) we observe an immediate and sustained increase of
pERK under HGF treatment in line with our simulation. Differences
in pERK level under simultaneous PAI-1 inhibition become most
apparent at late time points beyond 2 h, confirming the model
predictions of late impact of integrin signalling on ERK. Looking
at FAK as an up-stream effector of ERK (Sawhney et al., 2006),
we find similar activity for pFAK under HGF stimulation and
PAI-1 inhibition. We observe a strong, but late increase in FAK
activity as determined from phosphorylation of the Y925 site of
FAK (pFAK (Y925)). Simultaneous stimulation of HGF and PAI-1
inhibition decreases pFAK (Y925) when compared with HGF alone
(Supplementary Fig. S1). Immediate increase of pFAK (Y925)
suggests a possible role of FAK in the beginning of migration.
Previous work has shown two distinct phases in FAK involvement
for wound healing in rat keratinocytes (Providence and Higgins,
2004). There, the effect of PAI-1 inhibition became evident only
after 6 h and more into migration. Herein, we can possibly explain
the importance of FAK for prolonged migration through the time
sequential regulation of uPA/uPAR, integrin and EGFR signalling
pathways, which were predicted from network simulation and have
been — in part — experimentally validated. Further studies for
a better understanding of this complex pathway orchestration of
HGF-induced migration will be necessary, of course.
4
CONCLUSIONS
We have shown for the first time a comprehensive model for
HGF-induced keratinocyte migration. The model comprises two
time scales and incorporates various signalling pathway critically
involved in the initiation, sustaining and controlling of keratinocyte
migration. Being mostly compiled from prior knowledge and vast
literature, it will lend itself to rapid hypothesis testing of key points
of interference.
We are aware that the above model cannot capture the entire
complexity of this process. Most of the simulation results are
suggestive about the underlying process and further experiments
will need to be conducted to study the complex orchestration of
pathways leading to keratinocyte migration. However, our model
makes several important and experimentally testable predictions
i500
Boolean modelling of keratinocyte migration
about putative targets and time of intervention to control this process.
It underscores the temporal sequence of events from initial trigger
to execution, allowing for a fail-safe mechanism of migration under
wild-type and inhibition conditions. It is in line with previous
findings from literature and captures the short- and long-term
feedback regulation of protein signalling and gene expression. In
particular, the order of events becomes evident, how MET receptor
activity primes the cell for subsequent EGFR and integrin signalling,
leading sustained migration. Exchanging the order of stimulation
(first EGFR followed by MET receptor activation), keratinocytes
will not migrate, which is explained in the model and supported
by our first experimental results from the altered role of the FAK
proteins at late time points. Indeed, MET receptor deregulation is
a hallmark for cancer metastasis (Gherardi et al., 2012). If MET
is constitutively activated, it would need only additional EGFR
activity to induce cellular spread, according to the network model.
Apparently, this by-passes the fail-safe mechanism for cells to
migrate only within the correct context.Asynergistic action of EGFR
and MET have been found before (Brusevold et al., 2012; Zhou
et al., 2007), yet the causal relationship between them remained
unclear so far.
Beyond further experimental validation, the need for model
extensions
to
multiple
logical
states
or
even
continuous
representation of the variables using ordinary differential equation
approach is evident. Our current approach cannot account for
the multiple negative feedback regulations of either AKAP12 or
DUSP1. Such negative feedback seems to be a recurrent regulatory
motif (Becskei and Serrano, 2000; Blüthgen, 2010), and it should
be interesting to study its biological implications.
Taken together, the Boolean model approach lends itself to a better
mechanistic understanding of the process of keratinocyte migration,
wound healing, cancer metastasis and/or impaired wound healing
from stimulation to phenotype development and will help in the
prediction of cellular control targets in wild type and disease.
ACKNOWLEDGEMENT
A.S. acknowledges fruitful discussions with Jeremy Huard. M.B.,
H.B. and A.S. thank the MedSys consortium for fruitful discussions.
Funding: This work was supported by the Excellence Initiative of
the German Federal and State Governments. (FRIAS LifeNet to
H.B., M.B., S.K. and J.M.N.) and by the German Federal Ministry
of Education and Research (MedSys, Chronic Wounds; BMBF
0315401B, to A.S.).
Conflict of Interest: none declared.
REFERENCES
Albert,R. and Othmer,H.G. (2003) The topology of the regulatory interactions predicts
the expression pattern of the segment polarity genes in drosophila melanogaster.
J. Theor. Biol., 223, 1–18.
Becskei,A. and Serrano,L. (2000) Engineering stability in gene networks by
autoregulation. Nature, 405, 590–593.
Birchmeier,C. et al. (2003) Met, metastasis, motility and more. Nat. Rev. Mol. Cell
Biol., 4, 915–925.
Blüthgen,N. (2010) Transcriptional feedbacks in mammalian signal transduction
pathways facilitate rapid and reliable protein induction. Mol. Biosyst., 6, 1277–1284.
Bottaro,D.P. et al. (1991) Identification of the hepatocyte growth factor receptor as the
c-met proto-oncogene product. Science, 251, 802–804.
Brinkmann,V. et al. (1995) Hepatocyte growth factor/scatter factor induces a variety
of tissue-specific morphogenic programs in epithelial cells. J. Cell. Biol., 131,
1573–1586.
Brusevold,I.J. et al. (2012) Migration induced by epidermal and hepatocyte growth
factors in oral squamous carcinoma cells in vitro: role of mek/erk, p38 and pi-3
kinase/akt. J. Oral Pathol. Med. 2012 Mar 13. doi: 10.1111/j.1600-0714.2012.01139
Busch,H. et al. (2008) Gene network dynamics controlling keratinocyte migration. Mol.
Syst. Biol., 4, 199.
Chaves,M. et al. (2005) Robustness and fragility of boolean models for genetic
regulatory networks. J. Theor. Biol., 235, 431–449.
Cho,S.Y. and Klemke,R.L. (2000) Extracellular-regulated kinase activation and cas/crk
coupling regulate cell migration and suppress apoptosis during invasion of the
extracellular matrix. J. Cell. Biol., 149, 223–236.
Clague,M.J. (2011) Met receptor: a moving target. Sci. Signal., 4, pe40.
Gelman,I.H. (2010) Emerging roles for ssecks/gravin/akap12 in the control of cell
proliferation, cancer malignancy, and barriergenesis. Genes Cancer, 1, 1147–1156.
Gherardi,E. et al. (2012) Targeting met in cancer: rationale and progress. Nat. Rev.
Cancer, 12, 89–103.
Heng,M.C.Y. (2011) Wound healing in adult skin: aiming for perfect regeneration. Int.
J. Dermatol., 50, 1058–1066.
Hudson,L.G. and McCawley,L.J. (1998) Contributions of the epidermal growth factor
receptor to keratinocyte motility. Microsc. Res. Tech., 43, 444–455.
Hunt,T.K. et al. (2000) Physiology of wound healing. Adv. Skin Wound Care, 13, 6–11.
Jaakkola,P. et al. (1998) Wound reepithelialization activates a growth factor-responsive
enhancer in migrating keratinocytes. FASEB J., 12, 959–969.
Jeffers,M. et al. (1996) Autocrine hepatocyte growth factor/scatter factor-met signaling
induces transformation and the invasive/metastastic phenotype in c127 cells.
Oncogene, 13, 853–856.
Kain,K.H. and Klemke,R.L. (2001) Inhibition of cell migration by abl family
tyrosine kinases through uncoupling of crk-cas complexes. J. Biol. Chem., 276,
16185–16192.
Klemke,R.L. et al. (1997) Regulation of cell motility by mitogen-activated protein
kinase. J. Cell. Biol., 137, 481–492.
Legewie,S. et al. (2008) Recurrent design patterns in the feedback regulation of the
mammalian signalling network. Mol. Syst. Biol., 4, 190.
Medico,E. et al. (1996) The tyrosine kinase receptors ron and sea control “scattering”
and morphogenesis of liver progenitor cells in vitro. Mol. Biol. Cell., 7, 495–504.
Mendoza,L. (2006) A network model for the control of the differentiation process in th
cells. Biosystems, 84, 101–114.
Mendoza,L. et al. (1999) Genetic control of flower morphogenesis in Arabidopsis
thaliana: a logical analysis. Bioinformatics, 15, 593–606.
Mesecke,S. et al. (2011) Integration of activating and inhibitory receptor signaling by
regulated phosphorylation of vav1 in immune cells. Sci. Signal., 4, ra36.
Müssel,C. et al. (2010) Boolnet—an r package for generation, reconstruction and
analysis of boolean networks. Bioinformatics, 26, 1378–1380.
Muyderman,H. et al. (2001) Activation of beta-adrenoceptors opens calcium-activated
potassium channels in astroglial cells. Neurochem. Int., 38, 269–276.
Pastore,S. et al. (2008) The epidermal growth factor receptor system in skin repair and
inflammation. J. Invest. Dermatol., 128, 1365–1374.
Providence,K.M. and Higgins,P.J. (2004) Pai-1 expression is required for epithelial cell
migration in two distinct phases of in vitro wound repair. J. Cell. Physiol., 200,
297–308.
Samaga,R. et al. (2009) The logic of egfr/erbb signaling: theoretical properties and
analysis of high-throughput data. PLoS Comput. Biol., 5, e1000438.
Sawhney,R.S. et al. (2006) Integrin alpha2-mediated erk and calpain activation play
a critical role in cell adhesion and motility via focal adhesion kinase signaling:
identification of a novel signaling pathway. J. Biol. Chem., 281, 8497–8510.
Schäfer,M. and Werner,S. (2008) Cancer as an overhealing wound: an old hypothesis
revisited. Nat. Rev. Mol. Cell. Biol., 9, 628–638.
Schlatter,R. et al. (2009) On/off and beyond–a boolean model of apoptosis. PLoS
Comput. Biol., 5, e1000595.
Schnickmann,S. et al. (2009) Ap-1-controlled hepatocyte growth factor activation
promotes keratinocyte migration via ceacam1 and urokinase plasminogen
activator/urokinase plasminogen receptor. J. Invest. Dermatol., 129, 1140–1148.
Tsuboi,R. et al. (1993) Keratinocyte growth factor (fgf-7) stimulates migration and
plasminogen activator activity of normal human keratinocytes. J. Invest. Dermatol.,
101, 49–53.
Werner,S. et al. (2007) Keratinocytefibroblast interactions in wound healing. J. Invest.
Dermatol., 127, 998–1008.
Xue,M. et al. (2007) Protein c is an autocrine growth factor for human skin
keratinocytes. J. Biol. Chem., 282, 13610–13616.
Zhou,H.Y. et al. (2007) Synergistic effects of epidermal growth factor and hepatocyte
growth factor on human ovarian cancer cell invasion and migration: role of
extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase.
Endocrinology, 148, 5195–5208.
i501
|
22962472
|
EGFR = ( HBEGF ) OR ( EGF )
CDKN2A = ( Elk1 ) OR ( ETS )
DAG = ( PLC_g )
PAK3 = ( Cdc42_Rac1 )
uPA = ( uPAR )
Mkk4 = ( Mekk4 ) OR ( Mekk1 ) OR ( MLK3 )
p38 = ( ( Mkk3 AND ( ( ( Mkk6 ) ) ) ) AND NOT ( DUSP1 ) ) OR ( PAK1 )
PTGS2 = ( ATF2 ) OR ( cFOS AND ( ( ( cJUN ) ) ) )
JNK = ( Mekk7 AND ( ( ( Mkk4 ) ) ) ) OR ( PAK2 )
Mekk7 = ( Mekk1 )
CDKN1A = ( STAT3 )
Integrins = ( ECM )
CREB = ( RSK )
PAK2 = ( Cdc42_Rac1 )
Akt = ( ( PI3K ) AND NOT ( PTEN ) )
CCL20 = ( Erk )
RSK = ( Erk )
DOCK180 = ( CRKL )
SOS = ( Grb2 )
Mkk3 = ( MLK3 )
CyclinD = ( Elk1 ) OR ( ATF2 )
EGR1 = ( Erk )
Erk = ( Mek )
IP3 = ( PLC_g )
Mekk4 = ( Cdc42_Rac1 )
Proliferation = ( CDK2 AND ( ( ( CyclinD ) ) ) )
Plasmin = ( uPA AND ( ( ( PAI-1 ) ) ) )
Ca = ( IP3 )
AP1 = ( cFOS AND ( ( ( cJUN ) ) ) )
CRKL = ( Grb2 )
Ras = ( SOS )
STAT3 = ( Erk )
CellMigration = ( IL8 AND ( ( ( PTGS2 AND CTGF AND CCL20 ) ) ) )
PAK1 = ( Cdc42_Rac1 )
Cdc42_Rac1 = ( Akt AND ( ( ( Ras AND DOCK180 ) ) ) )
Mek = ( Raf ) OR ( Mekk1 )
PLC_g = ( EGFR ) OR ( MET )
Mekk1 = ( Cdc42_Rac1 )
Fak = ( ( Integrins AND ( ( ( Rap1 ) ) ) ) AND NOT ( PTEN ) )
Elk1 = ( JNK ) OR ( Erk )
PI3K = ( EGFR AND ( ( ( Fak ) ) ) ) OR ( MET )
cMYC = ( Erk )
Mkk6 = ( MLK3 )
uPAR = ( AP1 )
ECM = ( MMP1_10 )
cFOS = ( Erk )
cJUN = ( JNK AND ( ( ( p38 ) ) ) )
MLK3 = ( Cdc42_Rac1 )
Grb2 = ( Shc )
MET = ( HGF )
Raf = ( Ras AND ( ( ( PKC AND PAK3 ) ) ) )
HBEGF = ( p38 ) OR ( Erk )
Shc = ( Fak ) OR ( MET ) OR ( EGFR )
Rap1 = ( C3G )
PKC = ( ( DAG AND ( ( ( Ca ) ) ) ) AND NOT ( AKAP12 ) )
IL8 = ( p38 ) OR ( Erk )
CTGF = ( p38 ) OR ( Erk )
ATF2 = ( JNK AND ( ( ( p38 ) ) ) )
CDK2 = ( CyclinD AND ( ( ( NOT CDKN2A ) ) OR ( ( NOT CDKN1A ) ) ) )
ETS = ( Erk )
MMP1_10 = ( Plasmin )
C3G = ( CRKL )
|
Ergodic Sets as Cell Phenotype of Budding Yeast Cell
Cycle
Robert G. Todd*, Toma´sˇ Helikar
Department of Mathematics, University of Nebraska at Omaha, Omaha, Nebraska, United States of America
Abstract
It has been suggested that irreducible sets of states in Probabilistic Boolean Networks correspond to cellular phenotype. In
this study, we identify such sets of states for each phase of the budding yeast cell cycle. We find that these ‘‘ergodic sets’’
underly the cyclin activity levels during each phase of the cell cycle. Our results compare to the observations made in
several laboratory experiments as well as the results of differential equation models. Dynamical studies of this model: (i)
indicate that under stochastic external signals the continuous oscillating waves of cyclin activity and the opposing waves of
CKIs emerge from the logic of a Boolean-based regulatory network without the need for specific biochemical/kinetic
parameters; (ii) suggest that the yeast cell cycle network is robust to the varying behavior of cell size (e.g., cell division under
nitrogen deprived conditions); (iii) suggest the irreversibility of the Start signal is a function of logic of the G1 regulon, and
changing the structure of the regulatory network can render start reversible.
Citation: Todd RG, Helikar T (2012) Ergodic Sets as Cell Phenotype of Budding Yeast Cell Cycle. PLoS ONE 7(10): e45780. doi:10.1371/journal.pone.0045780
Editor: Jean Peccoud, Virginia Tech, United States of America
Received May 1, 2012; Accepted August 24, 2012; Published October 1, 2012
Copyright: 2012 Todd, Helikar. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: The authors have no support or funding to report.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: rtodd@unomaha.edu
Introduction
Complex network structures can be found across the biological
spectrum, and growing evidence indicates that these biochemical
networks have evolved to perform complex information processing
tasks in order for the cells to appropriately respond to the often
noisy and contradictory environmental cues [1]. While reduction-
ist techniques focus on the local interactions of biological
components, the systems approach aims at studying properties of
biological processes as a result of all components and their local
interactions working together [2].
A
wide spectrum
of modeling techniques
ranging from
continuous frameworks utilizing differential equations to discrete
(e.g., Boolean) techniques based on qualitative biological relation-
ships exist [3–5]. Each modeling technique is based on different
assumptions and hence comes with different advantages and
disadvantages.
Differential
equation
models can
depict the
dynamics of biological systems in great detail, but depend on
a large number of difficult-to-obtain biological (kinetic) parame-
ters. On the other hand, discrete modeling frameworks, namely
Boolean networks, are qualitative and parameter-free, which
makes them more suitable to study the dynamics of large-scale
systems for which these parameters are not available. Further-
more, probabilistic Boolean networks (PBN) enhance the discrete
framework by allowing for uncertainty and stochasticity (e.g.,
[6,7]).
It has been proposed that the irreducible sets of states (i.e.,
ergodic sets) of the corresponding Markov chain in probabilistic
Boolean network models (PBNs) are the stochastic analogue of the
limit cycle in a standard Boolean network, and should thus
represent cellular phenotype [8]. However, often PBNs with
perturbations are studied to include internal noise, rendering the
search for the irreducible sets trivial (as the whole state space
constitutes a single irreducible component). Furthermore, this
makes the determination of the limiting distribution of the
corresponding Markov chain and the interpretation of those
results in light of the biology challenging even for moderately sized
models [9–11].
Using the idea from [1] to introduce stochasticity to Boolean
models via control nodes, herein we determine and examine the
nature of ergodic sets of a regulatory network governing each
phase of the cell cycle of budding yeast, Saccharomyces cerevisiae. The
budding yeast cell cycle has been modeled previously using
Boolean approaches (e.g., [5,12,13]) and probabilistic Boolean
approaches (e.g., [7,14,15]). We expand on previous works by
considering each phase of the cell cycle as an individual evolving
system. The logic of the model used in this study was developed
from the description of the yeast cell cycle interactions given in
[12]. Using this model we show that as suggested in [8], irreducible
sets of states can correspond to cellular phenotype. This approach
enables us to model and visualize richer dynamical properties of
each phase and the cell cycle as a whole. In particular, we show
that under stochastic external signals the continuous oscillating
waves of cyclin activity and the opposing waves of CKIs that form
the cell cycle engine can emerge from the logic of a relatively
simple regulatory network without the need for specific bio-
chemical/kinetic parameters. Furthermore, by considering each
phase of the cell cycle as an individual system represented by an
‘‘ergodic set’’, we are able to more directly and precisely compare
the model dynamics with experimental studies. Specifically, results
of [16] as interpreted graphically at cyclebase.org reveal relatively
precise similarities. We also observe good agreement between our
oscillating cyclin activities and recently published analyses of cyclin
activities using fluorescent microscopy in [17]. The improved
approach to the modeling of the yeast cell cycle enables us to
visualize other qualitative features of the system: the secondary
PLOS ONE | www.plosone.org
1
October 2012 | Volume 7 | Issue 10 | e45780
activation of a number of G1-cyclins later in the cell cycle [16] and
the renewed reversibility of Start upon the removal of the Cln2-
SBF-Whi5 feedback loop [18]. We also capture the same
robustness to internal perturbations as described in [5], however
we extend this result and conclude that each phase of the yeast cell
cycle (and thus as the cell cycle as a whole) is robust in the face of
the variable behavior of the cell size. Within the model, this results
in the post-Start commitment to the cell cycle and the ability to
complete a single round of division under deprived nutrition
conditions [19].
Results
Modeling the Cell Cycle
The budding yeast cell cycle involves hundreds of species and
interactions [20]. In order to keep the mathematical analyses
manageable, we consider a much smaller network consisting of
some key players (see Methods for a narrative description of the cell
cycle). The logic of our network was constructed based on the
descriptions of the cell cycle interaction as given in section 3.1 of
[12], which is an expansion of the network found in [5]. All nodes,
the species they represent and the logic associated to each node,
are available in The Cell Collective (www.thecellcollective.org).
Figure 1 shows the static interaction graph of the model. The
model used in this study has four external inputs: cell size signal
(CSS) to model cell growth, the Start checkpoint (Start), the
budding (or morphogenic) checkpoint (BuddingCP) and the
spindle assembly checkpoint (SpindleCP). Each of these external
inputs plays a different role. First consider Start, BuddingCP, and
SpindleCP. Each external input is incorporated into the logic of
an internal node(s) so as to mimic the biological behavior of
a checkpoint. Activating one of these external inputs (setting it to 1)
indicates that the corresponding checkpoint has been satisfied. In
pre-Start cells, Cln3 cannot inhibit Whi5 nor can Cln2 be
activated unless the critical size threshold has been reached; hence
Start is integrated into the logic of Whi5 and Cln2 as follows: if
Start~0 then Whi5~1 and Cln2~0 [21–24]. The external input
BuddingCP corresponds to the correct formation of the bud neck
and the localization and subsequent degradation of Swe1 [25–27].
As such, we say that if BuddingCP~0 then Swe1~1. Lastly,
SpindleCP corresponds to the spindle assembly checkpoint which
modulates the activation of Cdc20: if SpindleCP~0 then
Cdc20~0 [28,29].
Finally, CSS is a signal representing cell size. It is known that
cell size regulates the cell cycle via its correlation with Cln3 levels.
The mechanism governing this regulation involves a complex
network of biochemical interactions [30,31], and has been omitted
for simplicity. Unlike the external nodes representing cell cycle
checkpoints, which are binary in nature (either satisfied or not), the
CSS external input is inherently continuous (cell size varies
continuously over time). To represent this continuous signal passed
from the cell and its environment to activate Cln3 at a given
moment, a probability (q) that CSS
is active is defined:
p(CSS~1)~q[(0,1). This signal is relative as p(CSS~1)~1
indicates Cln3 is receiving the strongest activation signal.
The cell cycle was modeled as a sequential activation of the
checkpoints. In other words, the pre-start or G1 phase was
modeled by setting all checkpoints to 0. The G1/S phase is
modeled by setting Start to 1. For the G2/M phase, BuddingCP is
set to 1; finally, the M/G1 phase is modeled via the activation of
the SpindleCP checkpoint. Hence, the cell cycle as a whole results
in a sequence of probabilistic Boolean control networks (PBCNs) (see the
Methods section for detailed discussion of PBCNs) as follows:
(CSS,Start,BuddingCP,SpindleCP)~
(q1(t),0,0,0)?(q2(t),1,0,0)?(q3(t),1,1,0)?(q4(t),1,1,1)
Call these PBCN1, PBCN2, PBCN3, PBCN4, where each qi(t) is
the control function governing CSS during each modeled phase.
The question is then: does each PBCN behave in accordance with
the phase assigned to it by the status of its checkpoints? In the next
section, the results of our analyses of the dynamics of these PBCNs
are presented.
Cyclin Activity Profiles Correspond to Ergodic Sets
To demonstrate how PBCNs can be used to visualize and
analyze the dynamics of biological systems, we first show that
ergodic sets correspond to cell phenotypes; i.e., cyclin activity
patterns of the individual cell cycle phases, in our case. Each PBCN
was analyzed, and ergodic sets were calculated. The question we
then asked: Do the cyclin activity functions of the individual
ergodic sets (and hence the modeled cell cycle phases) correspond
to the cyclin activity profiles during the cell cycle as seen in the
laboratory? In other words, does our model represent the
biological reality? In fact, the results of our analyses (discussed
below) indicate that the presented model accurately captures many
of the features of the species’ expected behavior (i.e., their activity
levels) during each cell cycle phase. In Figure 2 the ergodic sets
associated with each PBCN and the corresponding activity
functions of key cyclins as a function of q~CSS are summarized.
For each of the PBCNs exactly one ergodic set was found (ES1-4,
Figure 2A). For (ES1) the cyclin activity functions (column 1 in
Panel C) is consistent with pre-start G1 cells. The cyclin activity
functions of ergodic sets for PBCN2 and PBCN3 are consistent with
the G1/S and G2/M phase of the cell cycle, respectively (columns
2 and 3 in Figure 2A and B). Finally, during the last stage, the
cyclin activity functions of PBCN4 is consistent with M/G1 phase
when CSS is decreasing. That is, the cyclin dependent kinases
(e.g., Cln1{3, Clb2, etc.) deactivate while the cyclin kinase
inhibitors (e.g., Sic1 and Cdh1) reactivate. Thus we see that in fact
each PBCN does behave according the phase assigned to it by the
status of its checkpoints.
In order to model the dynamics of the cell cycle as a whole we
must consider how CSS is changing over time. Choosing an
appropriate qi(t) for i~1,2,3,4 as the control functions for CSS
for each corresponding phase we may see the behavior of each
species across the cell cycle as a whole. As time is arbitrary in our
model, we chose qi(t) to have the ith quarter of the unit interval as
its domain, and thus the modeled cell cycle to take one unit of
time. To organize the transition from one phase to the next we
suppose that if one concatenates those functions into a single
function q(t)~qi(t) if t[½i{1
4 , i
4 the overall behavior of CSS should
mimic cell size; i.e., it should grow for the majority of the cell cycle
and drop at the end. Furthermore, we assume that Cln3 peaks
when it is receiving the maximum signal. It has been shown that
the level of Cln3 rises and falls over the cell cycle and peaks
sometime during M phase [16,32]. Thus we let qi(t)~ 4
3 t with
domain ½i{1
4 , i
4 for i~1,2,3 and q4(t)~{4tz4 with domain
½3
4 ,1. (Note that the dynamical properties of the model are highly
robust to variations of the function, and thus our choice of the
control functions, as discussed in the Robustness section. ).
Consequently for each species, a piecewise function that governs
its activity across the cell cycle was constructed by composing each
node’s activity function with qi(t) during each corresponding
phase of the cell cycle. In Figure 3, one can see the control
Ergodic Sets for BYCC
PLOS ONE | www.plosone.org
2
October 2012 | Volume 7 | Issue 10 | e45780
function for CSS over the whole cell cycle on the left and the
corresponding activity of selected cyclins across the whole cell
cycle on the right. It is clear that we are able to reproduce the
general structure of the cell cycle. Cyclin kinase inhibitors are
active in G1, followed by their deactivation and the activation of
the G1 cyclins Clb5 and Cln2. Then the G1 cyclins deactivate and
Clb2 activates. Finally Clb2 deactivation correlates with Cdc20
activation as the cell progresses through M phase, and the
reactivation of the CKIs. (See Methods for a narrative description of
the cell cycle.) Direct comparison of the shape of the calculated
activity profiles to experimental studies in [16] (via cyclebase.org)
revealed a strong correspondence (Figure 4). Exceptions to this
correspondence with results from [16] were the dynamics of
Cdc14 and Cdh1. Our model predicts that Cdc14 is activated late
in the M phase (while inactive during the previous phases). This
behavior appears however to be consistent with another study that
suggested that Cdc14 activation occurs in late mitosis [33]. Also,
while the activation profile of Cdh1 predicted by our model
doesn’t agree with the results in [16], it appears to be consistent
with the activation profile described in Figure 2 in [4] (as well as all
other species common to each model). Thus not only does our
model’s results compare to laboratory results but also to the results
of a differential equation model. Note that the activity levels of
Whi5, Sic1, Cln2, Clb2, and Cdc20 also qualitatively correspond
to the combination of activity and localization measured in the cell
(see Figure 3 in [17]). Together, these data suggest that, in fact,
ergodic sets can model cell phenotypes. Furthermore, as visualized
in Figure 3 and Figure 4, a secondary peak of the G1 Cyclins (Cln2
and Clb5 in particular) was found as the cell transitions from
PBCN3 to PBCN4. In fact, this phenomenon was also observed in
the laboratory [16]. This is also a feature of the cyclin activity
profiles of the respective ergodic sets. Notice also that this peak is
not purely a result of the function that we chose for CSS. While
the shape of the peak may change, its existence is intrinsic to the
Figure 1. Regulatory Graph for Budding Yeast.
doi:10.1371/journal.pone.0045780.g001
Ergodic Sets for BYCC
PLOS ONE | www.plosone.org
3
October 2012 | Volume 7 | Issue 10 | e45780
logic of the network; that is, so long as CSS does not drop instantly
to zero when the spindle assembly checkpoint has been satisfied,
there will be some sharp rise and fall of Cln2 and Clb5.
Start, Irreversibility, and Commitment to the Cell Cycle
The irreversible nature of the Start signal was explored in [18]
by investigating the positive feedback loop that exists between
Cln2, SBFo (‘‘SMBF’’ in our model) and Whi5 (see Figure 1). As
can be seen in Figure 3B, the bi-stability of Cln2 is clearly
represented in the transition from the G1 ergodic set (ES1) where
Cln2 is inactive to the S-phase ergodic set (ES2) where Cln2 is
active. Notice that in PBCN1, the Whi5-Cln2-SMBF feedback
loop cannot be initiated, as Whi5 is active and Cln2 is inactive
until the Start signal has been received. On the other hand, in the
post-Start phase (i.e., PBCN2), Whi5 is now inhibiting itself as
a result of the Start-activated feedback loop. This suggests that the
feedback loop is inherent to the irreversibility of Start.
In the aforementioned work [18], the authors showed that
removing Cln2 from the feedback loop allows the reactivation of
Whi5 following an exogenous pulse of Cln2, rendering Start
reversible. To perform an analogous inquiry on our model, and to
investigate the role of the feedback loop, we eliminated Cln2 as an
upstream regulator of Whi5 and re-analyzed PBCN2. A single
ergodic set was found, whose cyclin activity profile is pictured in
Figure 5A. The functions from the activity profiles of ES1 and
ES2, that govern Cln2 when the feedback loop is present are
constant functions, and thus have no dependence on CSS. In
contrast, Cln2o and Whi5 activities are now a function of CSS
(Figure 5A). In other words, if the CSS stimulus is removed from
Cln3, Cln2 becomes inactive and Whi5 reactivates, indicating
a return to G1 phase and a renewed possibility of G1 arrest due to
mating pheromone [34]. The transition to S phase is now
reversible. Though the context of our model and what was done in
[18] are different, the result is the same – the irreversibility of the
G1/S transition is dependent on the positive feedback loop.
That the functions for the cyclins in the G1, S, and G2 phases
are constant has another implication for our model. Specifically,
once the Start signal has been received, the typical (oscillating)
activity profiles of the key cyclins will ensue even when stimulus of
Cln3 by cell size is incoherent, so long as the checkpoints are
satisfied. In other words, once the cell receives the Start signal, it
commits to a round of cell division.
Robustness
Robustness of biological systems is critical to the proper function
of processes such as the cell cycle. Within our modeling regime
Figure 2. Analysis of each PBCN. A) Ergodic sets (consisting of network states) for the individual PBCNs corresponding to individual cell cycle
phases. Each PBCN was constructed by changing the combination of satisfied checkpoints; the ‘‘activity’’ of the Cell Size Signal (CSS) external node is
defined by a probability q. Ergodic sets are visualized as nodes corresponding to network states (represented by their binary number +1) connected
by arrows illustrating the flow of these states. Red arrows correspond to q~1, blue arrows correspond to q = 0. Discussion of the individual ergodic
sets and their biological meaning can be found in the main text. B) Activity profiles (‘‘signatures’’) of cyclins during each modeled cell cycle phase. C)
Plots of cyclin activity functions as found by computing stationary distribution analytically using Maple 15, as discussed in the main text.
doi:10.1371/journal.pone.0045780.g002
Ergodic Sets for BYCC
PLOS ONE | www.plosone.org
4
October 2012 | Volume 7 | Issue 10 | e45780
noise is interpreted as the systems’ sensitivity to the control
function, and the robustness of the ergodic sets to random
perturbations, respectively. To consider the system’s sensitivity to
the control function, we considered the activity functions of the
ergodic sets. As noted in the previous section, the activity functions
governing most of the key species in the system are constant, and
hence independent of CSS during the first three phases of the cell
cycle. In particular, Cln2, Clb5, and Clb2, which drive bud
formation, DNA replication, and mitosis, respectively, have their
activities governed by constant functions. This indicates that so
long as the checkpoints are appropriately activated (i.e., the
environment is stable enough for the successful completion of the
current phase), the modeled cell will progress through the cell cycle
independently of CSS (i.e., cell size). Therefore our model is
robust to the variable behavior of the cell growth.
Furthermore, this is also consistent with the findings in [19] that
a cell deprived of nitrogen will proceed through one round of
division and arrest in G1. We modeled this scenario by removing
the cell size signal (CSS) right after Start has been satisfied.
Consistent with [35], as a control function we chose q1(t)~(4=3)t
for t[½0,1=4 and qi(t)~1=100 for t[( i{1
4 , i
4, i~1,2,3. The
dynamics of modeled cyclins are depicted in Figure 5B. During the
first three phases, the activities of the species are the same as the
normal cell cycle (Figure 2). The activities of the species during the
last phase are also consistent with a cell in the G1 phase. That is,
the modeled cell has completed a round of division and arrested in
G1. This may suggest that the phenomenon of completing a cell
cycle without appreciable growth is a consequence of the
robustness of the cell cycle to variable external environments,
and is inherent to the logic of the biological regulatory network
governing the yeast cell cycle.
In addition to being able to represent cellular phenotypes, the
calculated ergodic sets (and the number thereof) in the previous
section have another implication. Similar to attractors in Boolean
network, ergodic sets can provide insights into the robustness of
the modeled biological systems.
A standard approach to analyze robustness is to consider the
basins of attractions of each attractor and interpret its relative size
as a measure of stability (e.g., [5]). The concept of a basin of
attraction for an ergodic set in a PBCN is not well defined; this is
due to the fact that a random walk initiated from a single state in
the state space may arrive at different ergodic sets. However, each
of the PBCNs have a single ergodic set which means that any
perturbation will eventually return to the ergodic set (and can be
modeled by its associated cyclin activity functions). As such, we see
that the modeled G1, G1/S, G2/M and M/G1 phases of the cell
cycle are highly robust in the face of perturbations. Together, our
results suggest that each phase of the modeled cell cycle is robust as
well as the cell cycle as a whole.
Discussion
Results presented herein are twofold. First, as suggested in [8],
we show that it is possible to model cellular phenotype as ergodic
sets in the context of probabilistic Boolean control networks. In
contrast to previous works utilizing Boolean models, our approach
centers around understanding not only the cell cycle as a whole,
but also its individual phases. Specifically, we modeled the cell
cycle as a sequence of models, each representing an individual
phase in the cycle. This approach has significant implications as to
how the dynamics of the modeled cell cycle are interpreted and are
compared with experimental studies. Specifically, in previous
works the yeast cell cycle was modeled as a single system where the
phases were represented as transient states leading to a (fixed
point) attractor corresponding to the G1 phase [5], or as
consecutive states in a cyclic attractor [12,13]. Considering each
phase as an evolving system of its own enabled us to capture
continuous dynamics of key species during each phase and
compare them to laboratory studies. Modeling each phase
separately and transitioning between models via the activation of
checkpoints is also consistent with the biological observations that
it is not only kinase activity that causes phase transitions, but the
completion of each phases task [36].
Figure 3. The cell cycle in relative time. The left hand side depicts the control function for CSS along with the points in time where checkpoints
are activated. The right hand side depicts the concatenated activity profiles of the corresponding ergodic sets composed with CSS control function.
All species appear during each phase, though several my take on the same value, including 0.
doi:10.1371/journal.pone.0045780.g003
Ergodic Sets for BYCC
PLOS ONE | www.plosone.org
5
October 2012 | Volume 7 | Issue 10 | e45780
Figure 4. Comparison between our analytically calculated results (red) and the experimental results in [16] (green) via
cyclebase.org. Each node in the network may represent several species. In the case that a node represents more than one species its calculated
activity profile is compared to the experimental activation of the species to which it most clearly correlates. For example, the node Yhp1 in our model
represents the species YHP1 and YOX1. We thus compare the calculated profile of the node Yhp1 to YOX1, as they appear to have the best
correspondence. Numbered peaks and valleys identify our interpretation of the correlation between plots. The species corresponding to each node
can be found at thecellcollective.org.
doi:10.1371/journal.pone.0045780.g004
Ergodic Sets for BYCC
PLOS ONE | www.plosone.org
6
October 2012 | Volume 7 | Issue 10 | e45780
Similar to [5,12,13] we find that each phase of the cell cycle and
thus the cell cycle as a whole is robust as measured by basin size,
i.e. the existence of a single ergodic set for each phase. Stability of
the yeast cell cycle has also been considered in the framework of
probabilistic Boolean networks, concluding the cell cycle attractor
is robust to internal noise [7,15,37]. However, this approach is
incompatible with our goal of exploring the relevance of ergodic
sets as it renders the entire state space a single ergodic set.
Modeling extracellular signals as continuous variables (i.e., cell
size) allowed us show the stability of the yeast cell cycle network
under different choices of control functions, a question precluded
by previous modeling techniques. Lastly, taking the perspective of
qualitative activity introduced in [1] we are able to directly
incorporate the role of cell size into our model. The further
correlation of cell size with time also allows us to escape the
discrete time of other Boolean network models.
Evidence is increasing that biological processes possess complex
properties that emerge from the dynamics of the system working as
a whole (e.g., [1,30,38–41]). To better understand these emergent
properties, large-scale computational models of the complex
biological interactions will be needed. The size of the budding
yeast cell cycle network in this work is relatively small and makes
the analytic calculations manageable. Larger and more compre-
hensive models will be key in systems biology. For example,
understanding how the cell controls checkpoints via additional
regulatory network pathways, and how to incorporate this
understanding into current models is of paramount importance.
Thus the question of how to approach large networks is important
in extending these results to truly life-size scales. To deal with such
scales simulation techniques and software (such as The Cell
Collective; http://www.thecellcollective.org) will be an important
part of extending these results to large models.
Methods
Budding Yeast Cell Cycle
Newborn cells begin in the G1 phase of the cell cycle, where
they start growing. It isn’t until the cell reaches a critical size that
a round of division begins [42]. This transition point is referred to
as Start, and is irreversible; that is, once the Start signal is received,
the cell is no longer susceptible to G1 arrest due to mating
pheromone, and the cell has committed to a round of division,
[18,42,43]. The activity profile of the biochemical network
underlying
the
cell
cycle
during
the
initial
G1
phase
is
characterized by the increasing activity of the Cln3 cyclin in
response to the cell’s increasing size, and the activity of the cyclin
kinase inhibitor (CKI) Sic1 [42]. The transition to S phase occurs
once the critical size has been reached, i.e. Start has occurred, and
Cln1, 2 has become active and Sic1 has been inactivated. The
inactivity of Sic1 allows the activation of Clb5. Having transi-
tioned to S phase, the cells characteristic cyclin activity pattern is
the activity of Cln1, 2 and Clb5 and the inactivity of Sic1. During
S phase, Cln1, 2 allow bud and spindle-pole body formation, while
the activity of Clb5 allows DNA replication [18,23]. In G2 phase,
Clb2 (the primary mitotic cyclin) accumulates [44], and Swe1 is
degraded in the newly formed bud neck [25–27]. In fact, bud
formation (along with other nuclear events) constitutes another
quality control point: a morphogenic checkpoint [27]. The activity
of Clb2 is sustained into early M phase [44,45]. Thus one may say
that active Clb2 (and inactive Sic1) characterizes the G2/M phase
of the cell cycle. Further progression through M phase is governed
by another checkpoint: the spindle assembly checkpoint. Once the
chromosomes are correctly aligned on the mitotic spindle, Cdc20,
a co-factor of the ubiquitin ligase anaphase-promoting complex/
cyclosome (APC/C), is released from inhibition. The cell then will
progress through the rest of M phase and divide into a mother and
daughter cell in G1 phase, awaiting another round of division.
Figure 5. Irreversibility and commitment to cell cycle. A) Cln2 becomes inactive and Whi5 reactivates when CSS stimulation is removed. Thus
breaking Whi5-SMBF-Cln2 feedback loop makes Start reversible. B) Modeled cell cycle under nitrogen deprivation. CSS is linear during the G1 phase
and drops to.01 there after.
doi:10.1371/journal.pone.0045780.g005
Ergodic Sets for BYCC
PLOS ONE | www.plosone.org
7
October 2012 | Volume 7 | Issue 10 | e45780
Thus,
the
activation
and
deactivation
of
Cdc20
and
the
corresponding recovery of the Sic1 and Cdh1 and characterize
the M/G1 phase of the cell cycle [29,46]. It is this oscillating
activity of cyclin-dependent kinases that ‘‘act as the master
regulator for cell cycle progression’’ [47].
The complete model is freely available for download and further
modifications in The Cell Collective software at http://www.
thecellcollective.org; [48].
Modeling Framework
As noted in the Introduction section, the modeling framework
herein was suggested by [1]. The essential perspective of this
framework is to suppose that at every moment of time, our
biological system is being modeled by the stationary distribution of
an irreducible Markov chain, whose states are an irreducible
subset of the state space for a probabilistic Boolean network (which
itself is a reducible Markov chain).
Consider a collection of n nodes fx1, . . . xng, representing
biological entities, each taking a value in f0,1g, and n{m Boolean
functions fi : f0,1gn?f0,1g, i~1, . . . ,n{m, where the function fi
is the logical rule governing xmzi. Call the nodes xmz1, . . . ,xn
internal nodes and call nodes x1, . . . xm external inputs, as they are
not governed by a Boolean function. Decompose the state space of
the original n nodes as the direct sum f0,1gm+f0,1gn{m so that
for v+w[f0,1gm+f0,1gn{m v represents the state of the external
inputs and w represents the state of the internal nodes. Notice that
for each v[f0,1gm we may define Fv : f0,1gn{m?f0,1gn{m by
Fv(w)~(f1(v+w), . . . ,fn{m(v+w)) (we suppress the notation for
the standard inclusion of the direct sum). Thus we have defined
a family of 2m Boolean networks consisting of the internal nodes,
one for each vector in f0,1gm.
Suppose that to each external input xi we associate a function
qi(t) taking values in (0,1) with t[D, some arbitrary domain
representing time. Call qi(t) a control function. We suppose that
this probability represents the qualitative activation of the species
represented by xi at time t. Let t[D be fixed and consider the
probability
distribution
nt
on
f0,1gm
given
by
nt(v)~ Pm
i~1 qi(t)vi(1{qi(t))1{vi. Using this construct PBNt,
a probabilistic Boolean network where the probability that Fv is
chosen to update the network is nt(v). Abusing notation so that Fv
Figure 6. An example calculation. A) A diagram of a sample network with one external input. The logic of the internal nodes is represented with
Boolean truth tables. B) The state space associated with the network. Nodes are labeled by (bz1) where b is the binary number corresponding to the
activity of (N1,N2,N3,N4). Supposing that the probability that EI is active is q, the state space is traversed on dashed arrows with probability q and
solid arrows with probability 1{q. Nodes labeled with an underline constitute the ergodic set. C) On the left is the probabilistic transition matrix that
governs the system once it has reached the ergodic set. With the matrix is associated a unit modulus eigenvector that provides the invariant
distribution for the system. D) Each state of the ergodic set gives the activities of the internal nodes. Taking the sum of these binary vectors weighted
by the invariant distribution gives the likelihood that a particular node is active. Thus on the right of this expression is the activity function of each
node in the ergodic set. Note that the activity function is continuous for q[(0,1). E) In the left graph, the activity function of each node is plotted as
a function of q. In the middle graph, an arbitrary function for q (or the activity level of EI) is plotted as a function of time. In the graph on the right
side, the activities of the network nodes is plotted as a function of the composite function for EI in time (as designed in the middle graph).
doi:10.1371/journal.pone.0045780.g006
Ergodic Sets for BYCC
PLOS ONE | www.plosone.org
8
October 2012 | Volume 7 | Issue 10 | e45780
also stands for the state transition matrix of the associated Boolean
network, the state transition matrix of the corresponding Markov
chain is A
0
t~ P
v[f0,1gm nt(v)Fv. It is important to point out that we
may not assume that this Markov chain is irreducible, as we are
not considering any arbitrary perturbations of nodes. However,
within the state space f0,1gn{m there are recurrent communicat-
ing classes of states, and any random walk corresponding to this
Markov chain will arrive at one of these sets.
Suppose then that W~fw1, . . . ,wkg5f0,1gn{m is a recurrent
communicating class. Restricting the state transition matrix to W,
let At~A
0
tDW. At is an irreducible Markov chain. As we have no
guarantee that the resulting transition matrix is aperiodic we
consider pt, the stationary distribution on W associated to the
Markov chain At. We define the activity profile of the W at time t,
a recurrent communicating class of the original Markov chain, by
Pt~ Pk
i~1 pt(wi) wi. We then interpret the ith entry of Pt as the
qualitative activation of the species represented by node xi at time
t. It is important to note that the recurrent communicating classes
of PBNt are the same for all times t[D. (Thus, as above, we do not
need to index W by t.) This can be seen by understanding that the
recurrent communicating classes are determined by the semigroup
generated by the maps fFvDv[f0,1gmg, and not by the probabil-
ities associated to each Fv [49]. This is why we take care to assume
that each qi(t) takes values in (0,1)o since if at some time t,
qi(t)~0 or 1, then the semigroup associated to the Markov chain
has changed and thus the recurrent communicating classes at that
moment may be different. We will refer to this infinite family of
PBNs,
fPBNtDt[Dg,
associated
to
the
semigroup
S~fFvDv[f0,1gmg as a probabilistic Boolean control network.
Calculating Pt is aided by the fact that it can computed in two
steps. First we consider each qi as a formal variable instead as
a function of t. The matrix A is still stochastic, but its entries are
now 0’s, 1’s or polynomials in qi,i~1, . . . ,m. As such application
of the Perron-Frobenious theorem allows us to compute the
stationary distribution for this irreducible Markov chain as
a function which is continuous for all qi[(0,1). We call these the
activity functions for each ergodic set. We then compose these
functions with the control function for each qi rendering the
stationary distribution a function of t. Thus we have continuous
functions of t that give activity profile for the ergodic set at time t.
This procedure is demonstrated in a smaller example in Figure 6.
We used GAP (Groups, Algorithms, Programming) along with the
package Monoid written by James Mitchell in order to compute
the recurrent communicating classes for each PBCN. Maple 15
was used to compute the associated stationary distributions. For
further mathematical details see [49] and [50].
Model Construction via The Cell Collective
The Cell Collective (www.thecellcollective.org; [48]), is a collab-
orative modeling platform for large-scale biological systems. The
platform allows users to construct and simulate large-scale
computational models of various biological processes based on
qualitative interaction information. The platform’s Bio-Logic
Builder was used to create this yeast cell cycle models truth tables
by specifying the biological qualitative data (adopted from [12]).
The Cell Collective’s Knowledge Base component was also used to
catalog and annotate all biochemical/biological information for
the yeast cell cycle.
Acknowledgments
We thank Dr. Jim Rogers for his useful conversations and comments, and
Drs. Dora Matache and John Konvalina for their helpful comments on the
manuscript.
Author Contributions
Conceived and designed the experiments: RT TH. Performed the
experiments: RT TH. Analyzed the data: RT TH. Contributed
reagents/materials/analysis tools: RT TH. Wrote the paper: RT TH.
References
1. Helikar T, Konvalina J, Heidel J, Rogers JA (2008) Emergent decision-making
in biological signal transduction networks. Proc Natl Acad Sci U S A 105: 1913–
8.
2. Kitano H (2002) Systems biology: a brief overview. Science 295: 1662–4.
3. Gilbert D, Fuss H, Gu X, Orton R, Robinson S, et al. (2006) Computational
methodologies for modelling, analysis and simulation of signalling networks.
Brief Bioinform 7: 339–353.
4. Chen KC, Calzone L, Csikasz-Nagy A, Cross FR, Novak B, et al. (2004)
Integrative analysis of cell cycle control in budding yeast. Mol Biol Cell 15:
3841–3862.
5. Li F, Long T, Lu Y, Ouyang Q, Tang C (2004) The yeast cell-cycle network is
robustly designed. Proc Natl Acad Sci U S A 101: 4781–4786.
6. Kim S, Li H, Dougherty DR, Cao N, Chen Y, et al. (2002) Can markov chian
models mimic biological regulation? Journal of Biological Systesm 10: 337–357.
7. Zhang Y, Qian M, Ouyang Q, Deng M, Li F, et al. (2006) Stochastic model of
yeast cell cycle network. Physica D: Nonlinear Phenomena 219: 35–39.
8. Shmulevich I, Dougherty E, Kim S, Zhang W (2002) Probabilistic boolean
networks: a rule-based uncertainty model for gene regulatory networks.
Bioinformatics 18: 261–274.
9. Bruna M, Dougherty ER, Shmulevichc I (2005) Steady-state probabilities for
attractors in probabilistic boolean networks. Signal Processing 85.
10. Ching WK, Zhang S, Ng MK, Akutsu T (2007) An approximation method for
solving the steadystate probability distribution of probabilistic boolean networks.
Bioinformatics 23: 1511–1518.
11. Shmulevich I, Gluhosky I, Doughetry E, ZhangW (2003) Steady-state analysis of
genetic regulatory networks modelled by probabilitic boolean networks. Comp
Funct Genom 4: 601–608.
12. Irons D (2009) Logical analysis of the budding yeast cell cycle. Journal of
Theoretical Biology 257: 543–559.
13. Faure´ A, Naldi A, Lopez F, Chaouiya C, Ciliberto A, et al. (2009) Modular
logical modelling of the budding yeast cell cycle. Mol Biosyst 5: 1787–1796.
14. Zhang SQ, Ching WK, Ng MK, Akutsu T (2007) Simulation study in
probabilistic boolean network models for genetic regulatory networks. In-
ternational Journal of Data Mining and Bioinformatics 1.
15. Hasimota R, Stangi H, Higa CHA (2009) Budding yeast cell cycle modeled by
context sensitive probabilistic boolean network. In: Genomic Signal Processing
and Statistics, 2009. 1–4.
16. Cho RJ, Campbell MJ, Winzeler EA, Steinmetz L, Conway A, et al. (1998) A
genome-wide transcriptional analysis of the mitotic cell cycle. Mol Cell 2: 65–73.
17. Ball DA, Marchand J, Poulet M, Baumann WT, Chen KC, et al. (2011)
Oscillatory dynamics of cell cycle proteins in single yeast cells analyzed by
imaging cytometry. PLoS One 6.
18. Charvin G, Oikonomou C, Siggia ED, Cross FR (2009) Origin of irreversibility
of cell cycle start in budding yeast. PLos Biology 8.
19. Johnston GC, Pringle JR, Hartwell LH (1977) Coordination of growth with cell
division in the yeast saccharomyces cerevisiae. Exp Cell Res 105: 79–98.
20. Kaizu K, Ghosh S, Matsuoka Y, Moriya H, Shimizu-Yoshida Y, et al. (2010) A
comprehensive molecular interaction map of the budding yeast cell cycle. Mol
Syst Biol 6: 415–415.
21. Costanzo M, Nishikawa JL, Tang X, Millman JS, Schub O, et al. (2004) Cdk
activity antagonizes whi5, an inhibitor of g1/s transcription in yeast. Cell 117:
899–913.
22. Alberghina L, Rossi RL, Querin L, Wanke V, Vanoni M (2004) A cell sizer
network involving cln3 and far1 controls entrance into s phase in the mitotic
cycle of budding yeast. J Cell Biol 167: 433–43.
23. Wittenberg C, Reed SI (2005) Cell cycle-dependent transcription in yeast:
promoters, transcription factors, and transcriptomes. Oncogene 24: 2746–55.
24. Ferrell J (2011) Simple rules for complex processes: New lessons from the
budding yeast cell cycle. Molecular Cell 43: 497–500.
25. Lew DJ (2003) The morphogenesis checkpoint: how yeast cells watch their
figures. Curr Opin Cell Biol 15: 648–653.
26. Hood-DeGrenier JK, Boulton CN, Lyo V (2007) Cytoplasmic clb2 is required
for timely inactivation of the mitotic inhibitor swe1 and normal bud
morphogenesis in saccharomyces cerevisiae. Curr Genet 51: 1–18.
27. Lee K, Asano S, Park J, Sakchaisri K, Erikson R (2005) Monitoring the cell cycle
by multi-kinasedependent regulation of swe1/wee1 in budding yeast. Cell Cycle
10: 1346–1349.
Ergodic Sets for BYCC
PLOS ONE | www.plosone.org
9
October 2012 | Volume 7 | Issue 10 | e45780
28. Hwang LH, Lau LF, Smith DL, Mistrot CA, Hardwick KG, et al. (1998)
Budding yeast cdc20: a target of the spindle checkpoint. Science 279: 1041–
1044.
29. Musacchio A, Salmon ED (2007) The spindle-assembly checkpoint in space and
time. Nat Rev Mol Cell Biol 8: 379–393.
30. Barberis M, Klipp E, Vanoni M, Alberghina L (2007) Cell size at s phase
initiation: An emergent property of the g1/s network. PloS Computations
Biology 3.
31. Schneider BL, Zhang J, Markwardt J, Tokiwa G, Volpe T, et al. (2004) Growth
rate and cell size modulate the synthesis of, and requirement for, g1-phase
cyclins at start. Mol Cell Biol 24: 10802–10813.
32. McInerny CJ, Partridge JF, Mikesell GE, Creemer DP, Breeden LL (1997) A
novel mcm1-dependent element in the swi4, cln3, cdc6, and cdc47 promoters
activates m/g1-specific transcription. Genes Dev 11: 1277–88.
33. Bardin AJ, Amon A (2001) Men and sin: what’s the difference? Nat Rev Mol
Cell Biol 2: 815–826.
34. Strickfaden SC, Winters MJ, Ben-Ari G, Lamson RE, Tyers M, et al. (2007) A
mechanism for cell-cycle regulation of map kinase signaling in a yeast
differentiation pathway. Cell 128: 519–531.
35. Gallego C, Garı´ E, Colomina N, Herrero E, Aldea M (1997) The cln3 cyclin is
down-regulated by translational repression and degradation during the g1 arrest
caused by nitrogen deprivation in budding yeast. EMBO J 16: 7196–7206.
36. Hunt T, Nasmyth K, Novak B (2011) The cell cycle. Philisophical Transaction
of the Royal Society B 366: 3494–3497.
37. Ge G, Qian H, Qian M (2008) Synchronized dynamics and non-equilibrium
steadystates in a stochastic yeast cell-cycle network. Mathematical Biosciences
211: 132–152.
38. Bhalla US, Iyengar R (1999) Emergent properties of networks of biological
signaling pathways. Science 283: 381–7.
39. Alberghina L, Ho¨fer T, Vanoni M (2009) Molecular networks and system-level
properties. J Biotechnol 144: 224–233.
40. Efroni S, Harel D, Cohen IR (2007) Emergent dynamics of thymocyte
development and lineage determination. PLoS Comput Biol 3: e13.
41. Weng G, Bhalla US, Iyengar R (1999) Complexity in biological signaling
systems. Science 284: 92–6.
42. Rupes I (2002) Checking cell size in yeast. Trends Genet 18: 479–485.
43. Hereford L, Hartwell L (1974) Sequential gene function in the initiation of
saccharomyces cerevisiae dna synthesis. J Mol Biol 84: 445–461.
44. Maher M, Cong F, Kindelberger D, Nasmyth K, Dalton S (1995) Cell cycle-
regulated transcription of the clb2 gene is dependent on mcm1 and a ternary
complex factor. Mol Cell Biol 15: 3129–3137.
45. Grandin N, Reed SI (1993) Differential function and expression of saccharo-
myces cerevisiae b-type cyclins in mitosis and meiosis. Mol Cell Biol 13: 2113–
2125.
46. Chen RH (2007) Dual inhibition of cdc20 by the spindle checkpoint. J Biomed
Sci 14: 475–479.
47. Uhlmann F, Bouchoux C, Lopez-Aviles S (2011) A quantitative model for cyclin-
dependent kinase control of the cell cycle: revisited. Philisophical Transaction of
the Royal Society B 366: 3572–3583.
48. Helikar T, Kowal B, McClenathan S, Bruckner M, Rowley T, et al. (2012) The
Cell Collective: Toward an open and collaborative approach to systems biology.
BMC systems biology 6: 96.
49. Hognas G (1977) Random semigroup acts on a finite set. J Austral Math Soc
(Series A) : 481–498.
50. Norris J (1997) Markov Chains. Cambridge University Press.
Ergodic Sets for BYCC
PLOS ONE | www.plosone.org
10
October 2012 | Volume 7 | Issue 10 | e45780
|
23049686
|
Cdc14 = ( MEN AND ( ( ( FEAR ) ) ) )
Whi5 = ( NOT ( ( Cln3 AND ( ( ( Start ) ) ) ) OR ( Cln2 AND ( ( ( Start ) ) ) ) ) ) OR NOT ( Start OR Cln2 OR Cln3 )
Swi5 = ( ( Cdc14 AND ( ( ( NOT Swi5 ) ) AND ( ( SFF ) ) ) ) AND NOT ( Clb2 ) )
Cdh1 = ( ( Cdc14 ) ) OR NOT ( Cdc14 OR Clb2 OR Cln2 OR Clb5 )
Sic1 = ( ( ( ( ( Swi5 ) AND NOT ( Clb2 AND ( ( ( NOT Cdc14 ) ) ) ) ) AND NOT ( Clb5 AND ( ( ( NOT Cdc14 ) ) ) ) ) AND NOT ( Cln2 AND ( ( ( NOT Cdc14 ) ) ) ) ) OR ( ( ( ( Sic1 ) AND NOT ( Clb2 AND ( ( ( NOT Cdc14 ) ) ) ) ) AND NOT ( Clb5 AND ( ( ( NOT Cdc14 ) ) ) ) ) AND NOT ( Cln2 AND ( ( ( NOT Cdc14 ) ) ) ) ) ) OR NOT ( Swi5 OR Cdc14 OR Sic1 OR Clb2 OR Clb5 OR Cln2 )
FEAR = ( Cdc20 )
Clb2 = ( ( ( ( ( SFF ) AND NOT ( Cdh1 AND ( ( ( Cdc20 ) ) ) ) ) AND NOT ( Cdc20 AND ( ( ( Cdh1 ) ) ) ) ) AND NOT ( Swe1 ) ) ) OR NOT ( Cdc20 OR SFF OR Cdh1 OR Sic1 OR Swe1 )
Cln3 = ( ( Size ) AND NOT ( Yhp1 ) )
Clb5 = ( ( ( SMBF ) AND NOT ( Sic1 ) ) AND NOT ( Cdc20 ) )
Yhp1 = ( SMBF )
Swe1 = ( NOT ( ( BuddingCP ) ) ) OR NOT ( BuddingCP )
MEN = ( FEAR AND ( ( ( Clb2 ) ) ) )
Cdc20 = ( SpindleCP AND ( ( ( Clb2 ) ) AND ( ( SFF ) ) ) )
Cln2 = ( SMBF AND ( ( ( Start ) ) ) )
SMBF = ( NOT ( ( Whi5 ) OR ( Clb2 ) ) ) OR NOT ( Clb2 OR Whi5 )
SFF = ( Clb2 )
|
A Boolean Model of the Cardiac Gene Regulatory
Network Determining First and Second Heart Field
Identity
Franziska Herrmann1,2,3., Alexander Groß1,3., Dao Zhou1, Hans A. Kestler1*, Michael Ku¨ hl2*
1 Research Group Bioinformatics and Systems Biology, Institute for Neural Information Processing, Ulm University, Ulm, Germany, 2 Institute for Biochemistry and
Molecular Biology, Ulm University, Ulm, Germany, 3 International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
Abstract
Two types of distinct cardiac progenitor cell populations can be identified during early heart development: the first heart
field (FHF) and second heart field (SHF) lineage that later form the mature heart. They can be characterized by differential
expression of transcription and signaling factors. These regulatory factors influence each other forming a gene regulatory
network. Here, we present a core gene regulatory network for early cardiac development based on published temporal and
spatial expression data of genes and their interactions. This gene regulatory network was implemented in a Boolean
computational model. Simulations reveal stable states within the network model, which correspond to the regulatory states
of the FHF and the SHF lineages. Furthermore, we are able to reproduce the expected temporal expression patterns of early
cardiac factors mimicking developmental progression. Additionally, simulations of knock-down experiments within our
model resemble published phenotypes of mutant mice. Consequently, this gene regulatory network retraces the early steps
and requirements of cardiogenic mesoderm determination in a way appropriate to enhance the understanding of heart
development.
Citation: Herrmann F, Groß A, Zhou D, Kestler HA, Ku¨hl M (2012) A Boolean Model of the Cardiac Gene Regulatory Network Determining First and Second Heart
Field Identity. PLoS ONE 7(10): e46798. doi:10.1371/journal.pone.0046798
Editor: Robert Dettman, Northwestern University, United States of America
Received April 16, 2012; Accepted September 10, 2012; Published October 2, 2012
Copyright: 2012 Herrmann et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: The work was supported by the German Federal Ministry of Education and Research within its joint research project SyStaR and by the International
Graduate School in Molecular Medicine Ulm, funded by the Excellence Initiative of the German Federal and State Governments. The funders had no role in study
design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: michael.kuehl@uni-ulm.de; hans.kestler@uni-ulm.de
. These authors contributed equally to this work.
Introduction
The heart is the first functional organ to develop in mammals.
After the end
of gastrulation, cardiogenic
progenitor cells
constitute the cardiac crescent in the anterior mesoderm of the
murine embryo. At this stage the cardiogenic mesoderm splits
from a common cardiovascular progenitor cell population [1,2]
into two areas of differential gene expression: the so-called first
heart field (FHF) and the second heart field (SHF). Cells of the
FHF build the primary heart tube and later mainly contribute to
the left ventricle, most of the atria and provide a minority of cells
of the right ventricle. Cells of the SHF mainly contribute to the
right ventricle, the outflow tract and the atria [3,4].
Underlying regulatory factors control these differentiation
processes. The induction of mesoderm depends on canonical
Wnt
signaling
[5].
After
mesoderm
formation
cardiogenic
precursor cells are characterized by the expression of the
transcription factor Mesp1 [6]. Endodermal signals such as
Bmp2 were also described as being crucial for cardiogenesis
[7,8,9]. These signals activate a variety of transcription factors of
the cardiogenic mesoderm like Nkx2.5 or GATA factors [7,8,10].
Some of the cardiac transcription factors can be assigned to one of
the two heart fields. The transcription factors Isl1, Foxc1/2, Tbx1
and the ligand Fgf8 determine the area of the SHF, while the
transcription
factor
Tbx5
is
only
expressed
in
the
FHF
[11,12,13,14]. It is thought that intrinsic wiring among these
cardiac factors determines the progression of cardiac differentia-
tion and the division into subdomains of differential gene
expression. Heart development can severely be impaired in case
a regulatory factor of cardiogenesis is missing. Several studies
analyzed specific interactions within gene regulation of early
mammalian heart development using knock out or knock down
approaches of individual factors. A deeper understanding of the
cardiac gene regulatory network requires the implementation of
this network as a computational model and its subsequent analysis
by computational simulations.
Expression of a gene is regulated by input signals given by
transcription factors binding to the regulatory region of the
gene. The strength of transcription, e.g. the amount of primary
transcript, can be depicted as a function depending on the
concentration of these regulatory transcription factors. This
function often follows a sigmoidal behaviour, which is governed
by cooperativity in a first stage and controlled by saturation at
later stages resulting in a switch-like behavior. This property
ensures defined levels of gene expression for a wide range of
concentration levels. This sigmoidal function of gene expression
can be approximated as a step function [15]. A common
approximation of the possible states of a gene is therefore to
consider a gene to be active or inactive [15]. These two states
PLOS ONE | www.plosone.org
1
October 2012 | Volume 7 | Issue 10 | e46798
of a gene correspond to a present and to an absent gene
product and can be encoded as Boolean logical values: true (1)
and false (0). Dependencies between genes, e.g. whether a
transcription factor acts as a transcriptional activator, repressor
or both, can then be captured by Boolean functions which map
the state of a gene regulatory network to a succeeding state.
These functions allow a Boolean model to exhibit dynamical
behavior in simulations. Boolean logic network models have
been used to model e.g. endomesodermal territories in the sea
urchin [16], the hrp regulon of Pseudomonas syringae [17], and the
bistable lac operon in E. coli [18]. Every Boolean model has a
finite number of states, as the state of each gene is represented
by one of two possible values. For k genes this results in 2k
possible state combinations. It follows, that starting a simulation
from an initial state and following synchronous state transitions
according to the Boolean functions, the model eventually ends
up in recurring states, a cycle. A degenerated cycle may consist
of a single Boolean state. These recurring states are called
attractors. They can correspond to observed expression profiles
or phenotypes in gene regulatory networks [19,20,21].
We here introduce a Boolean model for the early cardiac gene
regulatory network of the mouse, containing known core genes
required for cardiac development and FHF/SHF determination.
The model is based on published temporal and spatial expression
patterns of relevant transcription factors and growth factors and
includes known regulatory interactions taking into account
whether a transcription factor acts as an activator or repressor
on a given target gene. We are able to show that this
computational model is able to reproduce different states observed
during cardiac development. Model simulations demonstrate that
stable states of gene expression representing either the FHF or the
SHF are encoded within the wiring of gene interactions. Thereby,
we provide an insight into the functional properties of the cardiac
gene regulatory network. This will be an important basis for
further enlargements of the network and for in-silico predictions of
genetic interactions.
Results
A Gene Regulatory Network for Early Murine
Cardiogenesis
For constructing a gene regulatory network of early cardiac
development we collected published data. An overview of cardiac
genes and their interactions is provided in Figure 1 and Table S1.
The expression of genes and their interactions take place in a
temporal and spatial frame, as marked by colored boxes in
Figure 1. The network is characterized by early signaling events
during gastrulation resulting in cardiac specification and subse-
quent signaling activities at the cardiac crescent stage which
separate the cardiac progenitor cell population into the territories
of the FHF and the SHF lineages. Furthermore, signaling from the
endoderm was also included into our model.
Early canonical Wnt signaling is required for the activation of
the pan-mesodermal transcription factor Brachyury [5,22] and for
the cardiac specific expression of the transcription factor Mesp1
[10]. Mesp1 subsequently activates various cardiac factors,
initiating the cardiac crescent stage. Mesp1 upregulates the
expression of genes of both heart fields, e.g. Nkx2.5, GATA4,
Tbx1 and Tbx5 [8,10]. At the same time, Mesp1 inhibits expression
of genes of other developmental fates (not included into the
model), e.g. endodermal genes and the mesodermal gene Brachyury
which later supports posterior mesoderm formation and axial
development [10,23,24]. In the FHF, cardiac transcription factors
like Nkx2.5, GATAs and Tbx5 build an intertwined positive
feedback circuit to stabilize their expression. They activate
downstream regulatory genes like Hand genes or myocardin. Finally,
regulatory factors upregulate a set of differentiation genes. Genes
being specific for terminal differentiation such as myosin light chain
genes (MLC) or myosin heavy chain genes (MHC) code for structural
proteins and constitute the end of the developmental processes
described by the regulatory network (Figure 1, FHF area).
Similarly, a network of molecular interactions exists in the SHF
(Figure 1, SHF area). Furthermore, the endoderm has been shown
to influence cardiogenesis, especially by the signaling factors Bmp2
and Dkk1. In addition, in the heart looping stage the regulatory
factors Isl1, Tbx1 and Fgf8 are also expressed in the pharyngeal
endoderm. There, Shh activates Tbx1 through the Forkhead
transcription factor Foxa2. Shh might also signal to the SHF and
regulate gene expression during later stages [12].
A Boolean Model of the Cardiac Regulatory Network
We implemented the core cardiac genes and their interactions
(Fig. 2A) as a Boolean model. The interactions of the involved
genes and signaling factors were gained from published data and
are enlisted in detail in Table S2. The corresponding Boolean
functions are given in Figure 2B. This computational model
represents the core regulatory interactions of the gene regulatory
network of cardiogenesis as presented in Figure 1.
As introduced, cardiac development also depends on signals
derived at particular time points of development from other tissues
such as the endoderm and thus are not included in the core
cardiac gene regulory network. Those are represented in our
model by four genes: exogen BMP2 I, exogen BMP2 II, exogen CanWnt
I, exogen CanWnt II. Bmp2 signaling for example from the
endoderm is important for the induction of cardiogenic mesoderm.
In order to represent the required activation of Bmp2 at the
correct time point, the cardiac progenitor cell state, we modeled a
temporal delay by the two genes exogen BMP2 I and exogen
BMP2 II. Similarly, exogen CanWnt I and exogen CanWnt II are
used to activate canonical Wnt signaling at the cardiac crescent
state in the SHF. This represents the described canonical Wnt
signaling in the SHF at E7.5 [25].
Expectations of the Boolean Model
Simulations of the computational Boolean model are expected
to reproduce gene expression profiles as closely as possible and in
the same temporal sequence as they appear during cardiogenesis in
vivo. Therefore, the temporal and spatial expression patterns of
genes included in the model were collected from publications
(Table S3). According to the collected data, expected attractors
representing the FHF and SHF as well as the expectations for
transient states were defined (Figure 3). Furthermore, as genes are
stably expressed in a certain area the model should not only
reproduce a gene expression profile, but exhibit also stability of
these states. Thus, we expect the expression profiles of the FHF
and of the SHF to be represented in attractors of the network
model.
The expected gene expression profiles for a FHF attractor and
for a SHF attractor are given in Figure 3A and Figure 3B,
respectively. In the FHF the genes Bmp2, Nkx2.5, Gatas and Tbx5
are expected to be active, while genes which usually only appear in
the SHF are inactive. In the SHF canWnt, Tbx1, Fgf8, Foxc1/2,
Gatas, Nkx2.5 and Isl1 are active, while Tbx5 is expected to be
inactive. Figure 3C summarizes the initial states, transition states
and the attractors of the FHF and the SHF.
Boolean Model of Cardiac Regulatory Network
PLOS ONE | www.plosone.org
2
October 2012 | Volume 7 | Issue 10 | e46798
The Network Model Exhibits Stable States for the FHF
and the SHF and Reproduces Temporal Development
To analyze the cardiac network, we simulated all possible initial
state combinations (215 = 32768 states for all genes) with the
cardiac network model. This analysis detects all possible Boolean
attractors of the Boolean model (Figure 4A). Our simulations lead
to three attractors. One attractor appears only for 1% of the initial
states and contains no active cardiac genes. Another attractor
appears in 49% of the cases. In this state the genes Bmp2, Tbx5,
GATA and Nkx2.5 are active, while other genes are inactive. This
attractor resembles the gene expression in FHF cells (compare
with Figure 3A) and will be called the FHF attractor from now on.
An additional attractor appears for 50% of all initial states and
corresponds to the SHF gene expression state (as specified in
Figure 3B). We call this the SHF attractor. This finding indicates
that the wiring of the network determines gene expression in the
first and second heart field.
Next, we analyzed the temporal sequence of network states
leading to the FHF and the SHF attractors. For this purpose we
included relevant biological information to define an appropriate
initial setting of the network. At the beginning of gastrulation,
canonical Wnt signaling is active and initiates the determination of
the mesodermal and cardiac cell lineage [26]. None of the other
cardiac specific genes included in our network are active at that
time. Therefore, we defined an initial state in which only canonical
Wnt signaling is active for further simulations with our network
model (compare expectation for the initial state in Figure 3C).
Furthermore, both heart fields are induced by endodermal Bmp
signaling after the induction of mesoderm. We modeled this by the
external signals exogen BMP2 I and exogen BMP2 II which
initiate Bmp signaling at the appropriate time point.
The initial state with active cardiac canWnt and exogen
(endodermal) BMP2 leads to a FHF attractor (Figure 4B, left
panel). After gastrulation, the SHF receives additional canonical
Wnt signaling while the FHF is unaffected. Therefore, we
additionally specified this external signal by exogen CanWnt I
and II for the setup which re-activates canonical Wnt signaling at
the cardiac crescent state. The initial state with active canWnt,
exogen BMP2 and exogen CanWnt leads to a SHF attractor
(Figure 4B, right panel).
Setups giving rise to both FHF and SHF contain an initial state
with active canWnt signaling and active exogen BMP2 I. The
difference between both setups is the additional activation of
exogen CanWnt I in the setup leading to the SHF. The use of
external signals allows us to compare the intrinsic states of the
network during the simulations to a temporal developmental
process in the developing organism, which integrates signals from
non-cardiac tissues.
The state transitions of the network, which occur during the
simulation, can be compared to a temporal process in the
developing organism. From the initial state on (marked by time
point t = 1) we follow the state transitions towards the final
attractors for the two setups differing in exogen CanWnt I
activation (Figure 4B). There are three state transitions leading to
the FHF attractor. Following an active canonical Wnt signaling
Mesp1 and Dkk1 are expressed at time point t = 2. Transient
Mesp1 expression is described in cardiac precursor cells during
gastrulation [6,27,28]. Mesp1 activates a variety of cardiac
regulatory factors [8,29]. The network state at t = 2 of the
simulation resembles the gastrulation stage in vivo (compare transit
1 in Figure 3C). Cardiac regulatory genes Nkx2.5, GATAs, Isl1 and
Tbx5 are activated at t = 3 of the simulation. Active genes of this
state resemble the expression in the common cardiovascular
progenitor cell population where Isl1, Tbx5, GATAs and Nkx2.5
are present [1,2,30]. The expected expression for this state is
defined in transition state 2 in Figure 3C. After one additional
transition the FHF resembling attractor appears (Figure 4B,
without exogen CanWnt I, t = 4).
In the setup with active exogen CanWnt I, cardiac canonical
Wnt signaling is reactivated at the state of the common
cardiovascular progenitor cell (Figure 4B, with exogen CanWnt
Figure 1. Gene regulatory network during early murine cardiac development. The overview comprises published gene regulations in early
heart precursor cells, focussing on two areas with different gene expression, the first heart field (FHF) and the second heart field (SHF). A
differentiation of the two heart fields happens around E7.5. Signaling of the endoderm which influences cardiac progenitors was included in this
overview as well as early mesodermal signals. Genes are represented by their regulatory region and their transcriptional start site. Information from
other genes is processed within the regulatory region. The transcriptional start site of a gene indicates expression and influences gene transcription
of other genes. Arrow heads indicate activation and bar heads inhibition of gene transcription. Broken lines represent intercellular signaling with an
integrated signal transduction cascade.
doi:10.1371/journal.pone.0046798.g001
Boolean Model of Cardiac Regulatory Network
PLOS ONE | www.plosone.org
3
October 2012 | Volume 7 | Issue 10 | e46798
I, t = 3). States leading to the common cardiovascular progenitors
are identical in comparison to the setup without exogen CanWnt I.
Canonical Wnt signaling leads to the activation of SHF genes and
thereby to the inhibition of the FHF transcription factor Tbx5.
The SHF attractor is constituted at time points t = 4 and t = 5. In
summary, the tracing of state transitions in the simple Boolean
model substantiates the literature derived expectations.
To verify that canonical Wnt signaling is the signaling event that
drives cells towards a SHF fate, we eliminated the influence of
exogen CanWnt II in the model by keeping exogen CanWnt II = 0
throughout the simulation. This is in line with investigations by
Klaus et al. [31] and Lin et al. [25] which showed that Wnt/aˆ-
catenin dependent transcription is upregulated at E7.5 in the
second heart field. For both initial setups the network attains a
FHF attractor, passing the same transition states as for the
wildtype without exogen canWnt (Figure 4B, left side). It
demonstrates that in our network model cardiac canonical Wnt
signaling which is induced from non cardiac tissue is sufficient to
establish the SHF attractor.
The Boolean Model Reproduces Knock-out and
Overexpression Phenotypes
For most of the genes involved in the presented cardiac network,
knock-out mice have been studied with respect to cardiac
development. To analyze whether our model will produce
attractors comparable to knock-out phenotypes, the corresponding
gene was kept off at all time points of simulation.
Figure 2. Boolean model of the cardiac gene regulatory network. (A) Only genes included into the model and their regulations are shown.
Regulations are based on published data. (B) Boolean transition functions of the network in (A). All genes of the network are listed on the left side. A
new state for each gene is derived from the value of the Boolean function on the right side based on the preceding states of genes. Input variables
are combined by Boolean functions: ! = NO, | = OR or & = AND. Brackets determine the order of evaluation, starting with the innermost. Exogen BMP2
I+II: inputs for non cardiac BMP2 signaling; exogen CanWnt I+II: inputs for non cardiac canonical Wnt signaling.
doi:10.1371/journal.pone.0046798.g002
Boolean Model of Cardiac Regulatory Network
PLOS ONE | www.plosone.org
4
October 2012 | Volume 7 | Issue 10 | e46798
Without Wnt signaling no mesoderm is formed, which is the
prerequisite for cardiogenesis [5]. For this, we set canWnt = 0,
simulating the ablation of cardiac canonical Wnt signaling
(Figure 5A). In the simulations of both setups Mesp1 is not
activated resulting in absence of cardiac mesoderm at time t = 2
and thus further cardiac factors are not activated. The predictions
of the model simulations are consistent with knock-out experi-
ments in mice, where upon ablation of canonical Wnt signaling no
mesoderm forms [5]. We also simulated overexpression of cardiac
canonical Wnt signaling by constantly activating Wnt signaling
(canWnt = 1) (Figure 5B). It is known that a sequence of different
levels of canonical Wnt signaling are required for proper heart
development [26]. In both initial setups, mesoderm is induced
upon Wnt overexpression as indicated by Mesp1 activation
(Figure 5B, t = 2). In the setup without exogen canonical Wnt
signaling, some of the cardiac genes are activated after expression
of Mesp1, but no defined heart field is established, leading to an
attractor without activated cardiac genes. In contrast, starting by
the setup with non-cardiac canonical Wnt signaling, the simulation
soon reaches the SHF attractor. In our model a FHF attractor is
not formed, while the SHF attractor is reached one time step
earlier. Conditional overexpression of b-catenin (a key transducer
of canonical Wnt signaling) disrupts primary heart tube formation,
a FHF derived structure and leads to an expanded Isl1 expression
in the SHF [25,31].
The mesodermal gene Mesp1 induces the expression of many
cardiac genes. In the mouse embryo Mesp2 can compensate for
some Mesp1 functions. Since Mesp2 is not an explicit part of the
network model, but merely is represented by Mesp1, model
simulations switching off Mesp1 (Mesp1 = 0) resembles a double
knock-out of Mesp1 and Mesp2 in the mouse embryo [28]. These
double knock-out mutant embryos do not form cardiac mesoderm.
In our model, switching of Mesp1 leads for the setup without
exogen canonical Wnt signaling to an attractor without any
expression of cardiac genes. For the setup with exogen CanWnt
signaling, no cardiac genes are active in the appropriate time
frame when a common cardiac progenitor cell should be
established (Figure 5C, with exogen CanWnt I, t = 3). If the time
window for differentiation of the cells passes, some other fate will
be adopted. In our simulations, the network reaches the SHF
attractor after a number of further state transitions due to a lack of
genes for alternative fates and a lack of further signaling input
from other tissues. Since our model does not exhibit cardiac gene
expression within two time steps past the gastrulation state, it is in
accordance with the reported in vivo results.
Dkk1 has been demonstrated to induce cardiac marker genes
together with Mesp1. Without Mesp1, Dkk1 inhibits their
expression [8]. The knock-out simulation of Dkk1 in the presented
cardiac gene regulatory network model exhibited a delay of
Nkx2.5 activation by one transition state. Besides this, the
attractors and state transitions are are identical in comparison to
the undisturbed network (Figure 5D). This simulation result
demonstrates that in our model Dkk1 is not important for the
correct specification and differentiation of cardiac mesoderm into
FHF and SHF lineages. As shown in [32], Dkk1 and Dkk2 double
mutant mice have no immediate effect on heart development.
Figure 3. Expected network states. Literature derived state descriptions are expected to appear in simulations, presented by a defined set of
genes to be active. In (A) a state defined as FHF state with the according gene activity and in (B) the expected SHF state are presented. Colored genes
are active, while gray genes are inactive. In (C) states are listed which are expected to appear during a simulation course. In the initial state of the
network which corresponds to early mesoderm development, only canonical Wnt signaling is expected to be present, while genes for differentiation
of cardiogenic precursor cells are still inactive. A simulation of the network model is expected to pass transition steps one and two. These correspond
to transient states of early cardiogenic mesoderm expressing Mesp1 and to the common cardiac progenitor cell population. Finally, the simulation is
expected to result in either the FHF or the SHF state. Genes which are expected to be expressed at a certain state are listed in green, and genes which
should not appear in a state are shown red. On the right side, the same information as in the table on the left is shown in a manner similar to the
results in Figures 4 and 5. In this table, white color indicates that no expectation for the gene activity is specified.
doi:10.1371/journal.pone.0046798.g003
Boolean Model of Cardiac Regulatory Network
PLOS ONE | www.plosone.org
5
October 2012 | Volume 7 | Issue 10 | e46798
Later, these double mutant mice exhibit defects in proliferation
and hypertrophy in the heart. These defects affect the heart later
than it is displayed in our network model and may result from a
later impact of Dkks on heart development. Furthermore, genes
regulating proliferation are not integrated into the network model
and can not be detected in our simulations.
The consistency between the biological phenotypes described in
the literature and our corresponding simulations demonstrate that
our Boolean model closely incorporates the molecular mechanisms
underlyingcardiacdevelopmentastheyhavebeeninvestigatedsofar.
Discussion
In the presented model of the cardiac gene regulatory network
we collected and integrated knowledge about major regulatory
factors required for heart development and their interactions. The
construction of a Boolean model of the cardiac regulatory network
allowed us to show that the interactions within the network lead to
stable regulatory states representing the FHF and SHF lineages at
E7.5 of murine development and indicates a robustness within the
network wiring.
Figure 4. Results of network model simulations. Gene activity for all genes of the model is presented at distinct network states. A green box
indicates activation whereas a red box denotes inactivation of a gene. (A) Summary of the analysis starting simulations from all possible initial states.
All runs resulted in one of the three attractors shown. 49% of the simulations resulted in an attractor for the FHF (indicated in purple), 50% in an
attractor mimicking the SHF state (indicated in blue) and 1% of the simulations yield an attractor without activation of core cardiac genes. (B)
Simulation of time courses from expected initial states of the cardiac gene regulatory network model. Initial state setups differ only in the activation
of canonical Wnt signaling at t = 3 (with exogen CanWnt I or without exogen CanWnt II). State transitions match expectations of intermediate state
expressions and end in the attractors for FHF and SHF lineages.
doi:10.1371/journal.pone.0046798.g004
Boolean Model of Cardiac Regulatory Network
PLOS ONE | www.plosone.org
6
October 2012 | Volume 7 | Issue 10 | e46798
Boolean Model of Cardiac Regulatory Network
PLOS ONE | www.plosone.org
7
October 2012 | Volume 7 | Issue 10 | e46798
The Boolean Model Describes Early Cardiac Development
We show that the computational model presented here is
sufficient to describe basic regulations of early heart development
in mice. Simulations of this model reproduce temporal behavior of
heart developmental processes reflecting important stages as the
canonical Wnt expression phases [26], a common cardiac
progenitor cell stage [1,2] and the FHF and SHF phenotypes
[3]. The model furthermore predicts the behaviour of the network
upon particular disturbance. Switching off initial canonical Wnt
signaling for example (Figure 5A; Wnt = 0) leads into a ’’no heart
field‘‘ attractor. This simulation result is consistent with the
complete ablation of b-catenin in mice, when mesoderm is not
formed [5]. Moreover, the transcription factor Mesp1 marks
cardiac progenitor cells. Upon switching off Mesp1 in our model,
either no heart field is developed or after activation of some
cardiac factors and six state transitions a SHF phenotype is
reached. After gastrulation, at about t = 3 or t = 4 cardiac genes
are not expressed in the simulation. At this stage usually a cardiac
fate is established. As Mesp1 expression, which is required for
cardiogenic induction, is not present and further network wiring
which could direct the network towards another attractor were not
included in the network the simulation ends in a SHF attractor.
Nevertheless, these results indicate that cardiac formation is
impaired upon Mesp1 ablation. This corresponds to the finding
that Mesp1 ablation in mouse leads to severe defects of the
formation of cardiogenic mesoderm. Upon ablation of Mesp1 and
Mesp2 no cardiac mesoderm is detected at all [27,28]. Further-
more, Dkk1 knock-out in our network model revealed no defect in
differentiation towards FHF and SHF fates and has barely an
impact on the activation of other cardiac factors. This is
comparable to Dkk1/Dkk2 double mutant mice, which do not
show defects in heart fate decision at the early stages of
development which the presented cardiac gene regulatory network
model covers [32]. Therefore, known in-vivo knock-out experi-
ments can be reproduced with the here presented mathematical
network model.
The stable state of the FHF is an example of intertwined
positive feedback regulation, as the factors involved activate
each other`s expression. The stabilization of the SHF is less
clear. In our computational model, it is mediated by canonical
Wnt signaling. Canonical Wnt signaling has been shown to act
in the SHF from E7.5 [25]. A conditionally ablated b-catenin (a
mediator of canonical Wnt signaling) mutant mouse leads to
shortened
SHF
derived
right
ventricle
and
outflow
tract.
Furthermore, SHF specific gene expression, especially Isl1, is
diminished in the outflow tract and in the splanchnic mesoderm
[25,31]. Analyses of a Mesp1-induced gain-of-function mutant
of b-catenin [31] shows that the formation of the linear heart
tube (usually promoted by FHF cells) was disrupted and the
expression
of
Isl1
was
expanded.
This
observation
is
in
agreement with the overexpression study in our Boolean model
(Figure 5B; Wnt = 1) which lacks the formation of a FHF state.
Together, these results indicate that canonical Wnt signaling is a
major regulator of SHF identity.
Wiring within the Network Results in Robustness
Our analyses demonstrate an important property of the network
wiring: robustness. Robustness ensures that aberrations in the
temporal appearance or in amounts of transcription factor
expression between cells do not alter the cell’s next stable
regulatory state.
Our simulations show that there are stable states of the network,
which resemble the gene expression patterns of the FHF and SHF,
respectively. This analysis investigated all possible initial states and
all transitions towards the attractors. The simulation of all possible
initial states has limited biological relevance, because most of these
initial states will not appear in a mouse embryo in vivo.
Nevertheless, the experiment shows that these attractors are an
intrinsic property of the Boolean network. The results can serve as
a measure of the stability against random fluctuations in gene
expression. Thus, most possible aberrations in states will eventually
lead to a FHF or SHF attractor through a self-stabilizing
mechanism contained in the wiring of the Boolean network.
Afterwards, for the progression of differentiation to a succeeding
stable regulatory state this robustness needs to be overruled e.g. by a
signalfromexternaltissue.Thissignalcanbetransmittedbysignaling
ligands for example. The cardiac network model e.g. contains
canonicalWntsignalingstartingtosignalfromnon-cardiactissueina
defined time frame. This signal affects the target cells to arrive at a
stable state, resembling the SHF. This state is only stable as long as
canonical Wnt signaling is present. If the canonical Wnt signal
discontinues, differentiation is directed to some other stable
regulatory state. Thus, robustness of a stable regulatory state is
required within the network, but also needs to be disabled by defined
signals in order to assure directed development.
Suitability of the Choosen Model to Simulate Cardiac
Development
Commonly, time dependent processes are modeled with
differential equations, which relate the change of a compound to
its value. These models describe networks of genes and require the
knowledge of concentrations for each involved species and kinetic
rates for each specified interaction [33,34,35]. Changes in time
and space further require additional parameters and can be
modeled using partial differential equations [36,37] or stochastic
approaches for low molecule numbers [38]. Differential equations
and stochastic approaches can accurately represent biological
systems if the quantitative data is available, which is rarely the case
for large systems [39]. In contrast, many approaches for modeling
gene regulatory networks are based on Boolean networks. Boolean
models were used for describing the cell cycle [40] or regulation of
segment polarity genes in Drosophila melanogaster [41]. Even
small Boolean networks with only a few genes show dynamic
behavior, which resembles experiments [42]. Another example for
a Boolean representation is the lac operon [18]. It demonstrates
activation and repression of gene transcription with bistability by
Boolean logic. These examples for Boolean models of gene
regulatory networks allow for a qualitative investigation and are
suitable for the analysis of larger networks [43,44].
Figure 5. Knock-down or overexpression simulations of canonical Wnt signaling, Mesp1 and Dkk1. For knock-down or overexpression a
gene is set to 0 or 1, respectively, throughout the simulation. Apart from that, the simulation is performed as in Figure 4B, starting from the defined
initial state and receiving signals as in the setup without non-cardiac canonical Wnt signaling or as in the setup with non-cardiac canonical Wnt
signaling. (A) Canonical Wnt signaling is switched off (canWnt = 0). This leads in both setups to attractors lacking active cardiac genes. (B) Upon
canonical Wnt overexpression (canWnt = 1), no FHF but a SHF forms. (C) If Mesp1 is switched off (Mesp1 = 0), no FHF builds. In the SHF setup, major
cardiac regulator genes are inactive after three state transitions. (D) Upon Dkk1 knock-out, attractors and transitions states stay the same. Activation
of Nkx2.5 is delayed by one step compared to the undisturbed simulation.
doi:10.1371/journal.pone.0046798.g005
Boolean Model of Cardiac Regulatory Network
PLOS ONE | www.plosone.org
8
October 2012 | Volume 7 | Issue 10 | e46798
The specified Boolean model represents states of single entities
by two values. This is a simplification of gene expression since in
vivo all factors can occur in more than two concentration levels.
The Boolean model is supported by the available data as in many
expression profiles discrete values are used describing either the
presence or absence of a transcription factor. The presence of
factors in embryonic tissue is mostly assessed by in-situ hybridisa-
tions or immunostainings and thereby provides qualitative data.
Additionally, the dependencies in the expression of factors are
often given just as positive or negative regulations. Some
interactions are measured by RT-PCR and stainings. This
qualitative data on states and relations can be translated without
many further considerations or interpretations into Boolean values
and functions. Nevertheless, the model integrates the different
findings on the cardiac gene regulatory network found in the
literature without inconsistencies. All input data used to model the
Boolean network is solely derived from literature. No estimations
of mechanisms and kinetic parameters or concentrations from the
available data were needed which would have been required for a
more complex model such as one based on differential equations.
The cardiac network model fulfills all conditions, which were
derived from various experimental studies found. Based on these
temporal and spatial expression and interaction data, the network
model attains a high validity. In order to fully validate the network
model, the expression pattern for all genes at all temporal stages in
the specific subset of cells would need to be determined.
Furthermore, knock-out or overexpression of these factors and
non cardiac signals in the respective developmental area would
help to test defined knock-out or overexpression situations of the
cardiac
regulatory
network.
These
data
compared
to
the
simulation of the Boolean network model would in the end fully
clarify whether the model completely displays and predicts
developmental processes or not. Only a limited amount of such
experimental data is available. Known results have been compared
to the results of the simulated Boolean model of the core cardiac
gene regulatory network and confirm our model.
Future Extensions of the Boolean Model
Beyond the regulations presented in the network, many more
factors, which are known to be expressed during cardiac
development, were not yet integrated into the model. Including
these factors and further interactions might refine the current
model. As new data on these relations become available the
computational model of the cardiac gene regulatory network will
be expanded to incorporate the new knowledge. This data might
be derived by multiple methods. These include time series of gene
expression data in cardiac tissue, prediction of transcription factor
binding sites in regulatory regions of cardiac genes, monitoring
whole genome transcription factor binding sites of factors of
interest by chromatin IP as well as by gene expression profiling
upon loss or gain of function of cardiac specific transcription
factors in geneticaly modified mice or differentiating murine ES
cells. Recently developed tools that allow for binarization of gene
expression data [45] and reverse engineering approaches [46,47]
will be of relevance for these purposes. Furthermore, logical
models using more than two states can provide an extension to
Boolean models [48,49]. In these models intermediate values for
gene states are needed requiring appropriate biological observa-
tions [50]. These and other extensions, like stochastic updates,
spatial dependency, or the restriction to certain subclasses of
models can then bridge the gaps as more data becomes available.
In summary, our here established initial model of the gene
regulatory network covering early cardiac development fulfills all
specified expectations and reproduces temporal and spatial gene
expression in early murine cardiogenesis. Thereby it helps to
deepen the picture of gene regulation dynamics during early
cardiogenesis including the consequences of misregulation as is
shown by the knock-out simulations. Furthermore, this Boolean
model will be the foundation for a growing gene regulation model
and further targeted experiments.
Materials and Methods
To generate the cardiac gene regulatory network, we collected
literature data about temporal and spatial expression of key genes
regulating cardiogenesis and their regulatory relations. These were
gained from experimental studies in mice, murine ES cells and in
two cases in a human cell line. A core set of genes was selected for
implementation as a Boolean model. Network components of the
model were chosen by their significance for heart development.
This is given by an expression between embryonic day 6.0 and 8.5
of heart development in mouse, and a sufficient amount of data
available concerning the regulation of expression. To concentrate
on core regulatory effects, genes integrated into the network
contain input as well as output relations. Furthermore, the
functional importance of genes involved has been shown by
impairment of cardiogenesis upon their knock-out in mice.
Network figures were drawn with Biotapestry (www.biotapestry.
org). Simulations of the Boolean network were performed with the
R package BoolNet [51] in R (www.r-project.org).
Our model contains four external signals (non cardiac BMP2
(exogen BMP2 I+II), non cardiac CanWnt (exogen CanWnt I+II).
They do not correspond to genes within the cardiac gene
regulatory network, but represent regulation input from non-
cardiac tissues as this has been described for cardiogenic cells. The
exogen BMP2 I and exogen BMP2 II represent a Bmp2 signal
from the endoderm while exogen CanWnt I and exogen CanWnt
II are required to reactivate cardiac canonical Wnt signaling in the
SHF at E7.5. Both external signals are represented by two non-
cardiac inputs in order to convey a temporal delay of two time
steps of the non-cardiac signals into the Boolean model of the
cardiac gene regulatory network.
Supporting Information
Table S1
Regulations of cardiac factors as depicted in
Figure 1 and their literature references.
(DOC)
Table S2
Regulatory interactions used in the cardiac
regulatory network model.
(PDF)
Table S3
Spatial and temporal expression pattern of
cardiac factors involved in the computational cardiac
network model.
(DOC)
Author Contributions
Conceived and designed the experiments: HAK MK. Wrote the paper: AG
FH MK HAK. Reviewed the literature and specified the model equations:
FH. Implemented the model: DZ. Carried out the simulations: AG DZ
FH. Analyzed the results, prepared the figures and supplements AG FH.
Boolean Model of Cardiac Regulatory Network
PLOS ONE | www.plosone.org
9
October 2012 | Volume 7 | Issue 10 | e46798
References
1. Moretti A, Caron L, Nakano A, Lam JT, Bernshausen A, et al. (2006)
Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle,
and endothelial cell diversification. Cell 127: 1151–1165.
2. Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, et al. (2008)
Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-
cell-derived population. Nature 453: 524–528.
3. Buckingham M, Meilhac S, Zaffran S (2005) Building the mammalian heart
from two sources of myocardial cells. Nature reviews Genetics 6: 826–835.
4. Laugwitz KL, Moretti A, Caron L, Nakano A, Chien KR (2008) Islet1
cardiovascular progenitors: a single source for heart lineages? Development 135:
193–205.
5. Liu P, Wakamiya M, Shea MJ, Albrecht U, Behringer RR, et al. (1999)
Requirement for Wnt3 in vertebrate axis formation. Nature genetics 22: 361–
365.
6. Kitajima S, Takagi A, Inoue T, Saga Y (2000) MesP1 and MesP2 are essential
for the development of cardiac mesoderm. Development 127: 3215–3226.
7. Liberatore CM, Searcy-Schrick RD, Vincent EB, Yutzey KE (2002) Nkx-2.5
gene induction in mice is mediated by a Smad consensus regulatory region.
Developmental biology 244: 243–256.
8. Lindsley RC, Gill JG, Murphy TL, Langer EM, Cai M, et al. (2008) Mesp1
coordinately regulates cardiovascular fate restriction and epithelial-mesenchymal
transition in differentiating ESCs. Cell stem cell 3: 55–68.
9. Zhang H, Bradley A (1996) Mice deficient for BMP2 are nonviable and have
defects in amnion/chorion and cardiac development. Development 122: 2977–
2986.
10. Bondue A, Lapouge G, Paulissen C, Semeraro C, Iacovino M, et al. (2008)
Mesp1 acts as a master regulator of multipotent cardiovascular progenitor
specification. Cell stem cell 3: 69–84.
11. Cai CL, Liang X, Shi Y, Chu PH, Pfaff SL, et al. (2003) Isl1 identifies a cardiac
progenitor population that proliferates prior to differentiation and contributes a
majority of cells to the heart. Developmental cell 5: 877–889.
12. Yamagishi H, Maeda J, Hu T, McAnally J, Conway SJ, et al. (2003) Tbx1 is
regulated by tissue-specific forkhead proteins through a common Sonic
hedgehog-responsive enhancer. Genes & development 17: 269–281.
13. Kelly RG, Brown NA, Buckingham ME (2001) The arterial pole of the mouse
heart forms from Fgf10-expressing cells in pharyngeal mesoderm. Developmen-
tal cell 1: 435–440.
14. Bruneau BG, Logan M, Davis N, Levi T, Tabin CJ, et al. (1999) Chamber-
specific cardiac expression of Tbx5 and heart defects in Holt-Oram syndrome.
Developmental biology 211: 100–108.
15. de Jong H (2002) Modeling and Simulation of Genetic Regulatory Systems: A
Literature Review. Journal of Computational Biology 9: 67–103.
16. Smith J, Theodoris C, Davidson EH (2007) A Gene Regulatory Network
Subcircuit Drives a Dynamic Pattern of Gene Expression. Science 318: 794–
797.
17. MacLean D, Studholme DJ (2010) A Boolean Model of the Pseudomonas
syringae hrp Regulon Predicts a Tightly Regulated System. PLoS ONE 5:
e9101.
18. Veliz-Cuba A, Stigler B (2011) Boolean Models Can Explain Bistability in the lac
Operon. Journal of Computational Biology 18: 783–794.
19. Thomas R, Kaufman M (2001) Multistationarity, the basis of cell differentiation
and memory. I. Structural conditions of multistationarity and other nontrivial
behavior. Chaos: An Interdisciplinary Journal of Nonlinear Science 11: 170–
179.
20. Thomas R, Kaufman M (2001) Multistationarity, the basis of cell differentiation
and memory. II. Logical analysis of regulatory networks in terms of feedback
circuits. Chaos: An Interdisciplinary Journal of Nonlinear Science 11: 180–195.
21. Kauffman SA (1993) The Origins of Order: Self Organization and Selection in
Evolution: Oxford University Press.
22. Arnold SJ, Stappert J, Bauer A, Kispert A, Herrmann BG, et al. (2000)
Brachyury is a target gene of the Wnt/beta-catenin signaling pathway.
Mechanisms of development 91: 249–258.
23. Kispert A, Herrmann BG (1994) Immunohistochemical analysis of the
Brachyury protein in wild-type and mutant mouse embryos. Dev Biol 161:
179–193.
24. Wilkinson DG, Bhatt S, Herrmann BG (1990) Expression pattern of the mouse
T gene and its role in mesoderm formation. Nature 343: 657–659.
25. Lin L, Cui L, Zhou W, Dufort D, Zhang X, et al. (2007) Beta-catenin directly
regulates Islet1 expression in cardiovascular progenitors and is required for
multiple aspects of cardiogenesis. PNAS 104: 9313–9318.
26. Gessert S, Ku¨hl M (2010) The multiple phases and faces of wnt signaling during
cardiac differentiation and development. Circulation research 107: 186–199.
27. Saga Y, Miyagawa-Tomita S, Takagi A, Kitajima S, Miyazaki J, et al. (1999)
MesP1 is expressed in the heart precursor cells and required for the formation of
a single heart tube. Development (Cambridge, England) 126: 3437–3447.
28. Saga Y, Kitajima S, Miyagawa-Tomita S (2000) Mesp1 expression is the earliest
sign of cardiovascular development. Trends in cardiovascular medicine 10: 345–
352.
29. David R, Brenner C, Stieber J, Schwarz F, Brunner S, et al. (2008) MesP1 drives
vertebrate cardiovascular differentiation through Dkk-1-mediated blockade of
Wnt-signalling. Nature cell biology 10: 338–345.
30. Kattman SJ, Huber TL, Keller GM (2006) Multipotent flk-1+ cardiovascular
progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth
muscle lineages. Developmental cell 11: 723–732.
31. Klaus A, Saga Y, Taketo MM, Tzahor E, Birchmeier W (2007) Distinct roles of
Wnt/beta-catenin and Bmp signaling during early cardiogenesis. PNAS 104:
18531–18536.
32. Phillips MD, Mukhopadhyay M, Poscablo C, Westphal H (2012) Dkk1 and
Dkk2 regulate epicardial specification during mouse heart development.
International Journal of Cardiology 150: 186–192.
33. Li S, Brazhnik P, Sobral B, Tyson JJ (2008) A Quantitative Study of the Division
Cycle of Caulobacter crescentus Stalked Cells. PLoS Comput Biol 4: e9.
34. Chen KC, Calzone L, Csikasz-Nagy A, Cross FR, Novak B, et al. (2004)
Integrative Analysis of Cell Cycle Control in Budding Yeast. Molecular Biology
of the Cell 15: 3841–3862.
35. Wawra C, Ku¨hl M, Kestler HA (2007) Extended analyses of the Wnt/beta-
catenin pathway: robustness and oscillatory behaviour. FEBS letters 581: 4043–
4048.
36. Kestler HA, Ku¨hl M (2011) Generating a Wnt switch: it’s all about the right
dosage. The Journal of cell biology 193: 431–433.
37. Turing AM (1952) The Chemical Basis of Morphogenesis. Philosophical
Transactions of the Royal Society of London Series B, Biological Sciences 237:
37–72.
38. Gillespie DT (1977) Exact Stochastic Simulation of Coupled Chemical
Reactions. The Journal of Physical Chemistry 81: 2340–2361.
39. Kestler HA, Ku¨hl M (2008) From individual Wnt pathways towards a Wnt
signalling network. Philosophical Transactions of the Royal Society of London
Series B, Biological sciences 363: 1333–1347.
40. Faure´ A, Naldi A, Chaouiya C, Thieffry D (2006) Dynamical analysis of a
generic Boolean model for the control of the mammalian cell cycle.
Bioinformatics 22: e124–e131.
41. Albert R, Othmer HG (2003) The topology of the regulatory interactions
predicts the expression pattern of the segment polarity genes in Drosophila
melanogaster. Journal of Theoretical Biology 223: 1–18.
42. Giacomantonio CE, Goodhill GJ (2010) A Boolean Model of the Gene
Regulatory Network Underlying Mammalian Cortical Area Development. PLoS
Comput Biol 6: e1000936.
43. Bornholdt S (2005) Systems biology. Less is more in modeling large genetic
networks. Science 310: 449–451.
44. Bornholdt S (2008) Boolean network models of cellular regulation: prospects and
limitations. Journal of the Royal Society, Interface 5 Suppl 1: S85–94.
45. Hopfensitz M, Mu¨ssel C, Wawra C, Maucher M, Ku¨hl M, et al. (2011)
Multiscale Binarization of Gene Expression Data for Reconstructing Boolean
Networks. IEEE/ACM Transactions on Computational Biology and Bioinfor-
matics 9: 487–498.
46. La¨hdesma¨ki H, Shmulevich I, Yli-Harja O (2003) On learning gene regulatory
networks under the Boolean network model. Machine Learning 52: 147–167.
47. Maucher M, Kracher B, Ku¨hl M, Kestler HA (2011) Inferring Boolean network
structure via correlation. Bioinformatics 27: 1529–1536.
48. Ghysen A, Thomas R (2003) The formation of sense organs in Drosophila: a
logical approach. BioEssays : news and reviews in molecular, cellular and
developmental biology 25: 802–807.
49. Espinosa-Soto C, Padilla-Longoria P, Alvarez-Buylla ER (2004) A Gene
Regulatory Network Model for Cell-Fate Determination during Arabidopsis
thaliana Flower Development That Is Robust and Recovers Experimental Gene
Expression Profiles. The Plant Cell 16: 2923–2939.
50. Kestler HA, Wawra C, Kracher B, Ku¨hl M (2008) Network modeling of signal
transduction: establishing the global view. BioEssays 30: 1110–1125.
51. Mu¨ssel C, Hopfensitz M, Kestler HA (2010) BoolNet - an R package for
generation, reconstruction and analysis of Boolean networks. Bioinformatics 26:
1378–1380.
Boolean Model of Cardiac Regulatory Network
PLOS ONE | www.plosone.org
10
October 2012 | Volume 7 | Issue 10 | e46798
|
23056457
|
Bmp2 = ( ( exogen_BMP2_II ) AND NOT ( canWnt ) )
GATAs = ( Nkx2_5 ) OR ( Tbx5 ) OR ( Mesp1 )
Isl1 = ( Mesp1 ) OR ( Tbx1 ) OR ( Fgf8 ) OR ( canWnt AND ( ( ( exogen_canWnt_II ) ) ) )
exogen_canWnt_II = ( exogen_CanWnt_I )
canWnt = ( exogen_canWnt_II )
Nkx2_5 = ( Tbx1 ) OR ( Mesp1 AND ( ( ( Dkk1 ) ) ) ) OR ( Bmp2 AND ( ( ( GATAs ) ) ) ) OR ( Tbx5 ) OR ( Isl1 AND ( ( ( GATAs ) ) ) )
Tbx1 = ( Foxc1_2 )
exogen_BMP2_II = ( exogen_BMP2_I )
Foxc1_2 = ( canWnt AND ( ( ( exogen_canWnt_II ) ) ) )
Mesp1 = ( ( canWnt ) AND NOT ( exogen_BMP2_II ) )
Tbx5 = ( ( ( ( Nkx2_5 ) AND NOT ( Dkk1 AND ( ( ( NOT Mesp1 AND NOT Tbx5 ) ) ) ) ) AND NOT ( Tbx1 ) ) AND NOT ( canWnt ) ) OR ( ( ( ( Tbx5 ) AND NOT ( Dkk1 AND ( ( ( NOT Mesp1 AND NOT Tbx5 ) ) ) ) ) AND NOT ( Tbx1 ) ) AND NOT ( canWnt ) ) OR ( ( ( ( Mesp1 ) AND NOT ( Dkk1 AND ( ( ( NOT Mesp1 AND NOT Tbx5 ) ) ) ) ) AND NOT ( Tbx1 ) ) AND NOT ( canWnt ) )
Dkk1 = ( Mesp1 ) OR ( ( canWnt ) AND NOT ( exogen_BMP2_II ) )
Fgf8 = ( ( Foxc1_2 ) AND NOT ( Mesp1 ) ) OR ( ( Tbx1 ) AND NOT ( Mesp1 ) )
exogen_CanWnt_I = ( exogen_CanWnt_I )
|
RESEARCH
Open Access
Boolean modeling and fault diagnosis in
oxidative stress response
Sriram Sridharan1, Ritwik Layek1, Aniruddha Datta1*, Jijayanagaram Venkatraj2
From IEEE International Workshop on Genomic Signal Processing and Statistics (GENSIPS) 2011
San Antonio, TX, USA. 4-6 December 2011
Abstract
Background: Oxidative stress is a consequence of normal and abnormal cellular metabolism and is linked to the
development of human diseases. The effective functioning of the pathway responding to oxidative stress protects
the cellular DNA against oxidative damage; conversely the failure of the oxidative stress response mechanism can
induce aberrant cellular behavior leading to diseases such as neurodegenerative disorders and cancer. Thus,
understanding the normal signaling present in oxidative stress response pathways and determining possible
signaling alterations leading to disease could provide us with useful pointers for therapeutic purposes. Using
knowledge of oxidative stress response pathways from the literature, we developed a Boolean network model whose
simulated behavior is consistent with earlier experimental observations from the literature. Concatenating the
oxidative stress response pathways with the PI3-Kinase-Akt pathway, the oxidative stress is linked to the phenotype of
apoptosis, once again through a Boolean network model. Furthermore, we present an approach for pinpointing
possible fault locations by using temporal variations in the oxidative stress input and observing the resulting
deviations in the apoptotic signature from the normally predicted pathway. Such an approach could potentially form
the basis for designing more effective combination therapies against complex diseases such as cancer.
Results: In this paper, we have developed a Boolean network model for the oxidative stress response. This model was
developed based on pathway information from the current literature pertaining to oxidative stress. Where applicable,
the behaviour predicted by the model is in agreement with experimental observations from the published literature.
We have also linked the oxidative stress response to the phenomenon of apoptosis via the PI3k/Akt pathway.
Conclusions: It is our hope that some of the additional predictions here, such as those pertaining to the
oscillatory behaviour of certain genes in the presence of oxidative stress, will be experimentally validated in the
near future. Of course, it should be pointed out that the theoretical procedure presented here for pinpointing fault
locations in a biological network with feedback will need to be further simplified before it can be even considered
for practical biological validation.
Introduction
The control of gene expression in eukaryotic organisms is
achieved via multivariate interactions between different
biological molecules such as proteins and DNA [1].
Consequently, in recent years, various genetic regulatory
network modeling approaches such as differential equa-
tions and their discrete-time counterparts, Bayesian
networks, Boolean networks (BNs) and their probabilistic
generalizations, the so-called probabilistic Boolean
networks (PBNs) [2] have been proposed for capturing the
holistic behavior of the relevant genes. Some of these
approaches such as differential equations involve finer
models and require a lot of data for inference while others
such as Boolean networks yield coarse models with lower
data requirements for model inference. On the other
hand, historically biologists have focused on experimen-
tally establishing marginal cause-effect relationships
between different pairs of genes, which when concatenated
* Correspondence: datta@ece.tamu.edu
1Texas A & M University, Electrical and Computer Engineering, College
Station, TX, 77843-3128, USA
Full list of author information is available at the end of the article
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
© 2012 Sridharan et al.; licensee BioMed Central Ltd. This is an open access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly cited.
together leads to what is known as pathway information.
Biological pathways are used by biologists to represent
complex interactions occurring at the molecular level
inside living cells [3]. Pathway diagrams describe how the
biological molecules interact to achieve their biological
function in the presence of appropriate stimuli [4]. At a
very simple level, biological pathways represent the graphi-
cal interactions between different molecules. However, as
already noted, the pathways give only a marginal picture
of the regulations (up-regulation or down-regulation) of
the different genes/RNAs/proteins by other genes/RNAs/
proteins.
The complexity of biological signaling and the preve-
lance of prior information in the form of pathway knowl-
edge demand that genetic regulatory network models
consistent with pathway information be developed.
Motivated by this, we developed an approach to generate
Boolean network models consistent with given pathway
information and applied it to studying the p53-mediated
DNA damage stress response [5]. In addition, we used a
signaling diagram of the MAP-Kinase pathways to predict
possible location(s) of the single signaling breakdowns,
based on the cancer-causing breakdown signature [6].
Moreover, we also made theoretical predictions of the effi-
cacy of different combination therapies involving six anti-
cancer drugs, which we plan to validate in the near future.
In this paper, we first develop a Boolean network model
consistent with oxidative stress response pathway informa-
tion from the biological literature. Thereafter this model is
linked with the PI3k/Akt pathway and the composite
model is used to pinpoint the possible fault locations
based on the observed deviations in the apoptotic signa-
ture over different time windows. The paper is organized
as follows. Section contains a brief general description of
Stress Response Pathways while Section presents a discus-
sion specific to the case of oxidative stress. The Boolean
network model for oxidative stress response is developed
in Section. The role of mitochondria as the site in a cell
where the oxidative stress is generated is discussed in Sec-
tion. In Section, we develop an integrated network linking
oxidative stress response to the phenomenon of apoptosis
via the PI3k/Akt pathways. Section presents an approach
for pinpointing fault locations in the integrated network
by observing the apoptotic signature in response to certain
test stress input sequences. Finally, Section contains some
concluding remarks.
Stress response pathways
Adaptive stress response pathways are the first responders
to chemical toxicity, radiation, and physical insults. The
different stress response pathways share a very similar
architecture. This architecture has three main compo-
nents: a transducer, a sensor and a transcription factor
(TF) [7]. The transcription factor (TF) is a DNA-binding
protein that interacts with the promoter regions of its
target genes via its canonical DNA-binding sites, known as
‘response elements’ (REs), to activate the expression of the
target genes. The sensor is a protein that physically inter-
acts with the transcription factor in the cytosol, sequester-
ing the transcription factor from the nucleus under
normal cellular conditions. In addition to its role in cyto-
plasmic sequestration of the TF, the sensor may direct TF
degradation, providing an additional layer of regulatory
control. The result of the sensor-TF complexation is to
maintain inactivity of the TF under normal cellular condi-
tions, while providing a mechanism that permits activation
in response to an appropriate insult to the cell. The trans-
ducer is an enzymatic protein, such as a kinase, that con-
veys a biochemical change from a signaling pathway
upstream of the sensor/TF complex in the event of cellular
stress. The transducer may directly modify the transcrip-
tion factor, providing the activating signal or modify the
sensor which in turn, destabilizes the sensor/TF complex.
Liberated, stabilized, and activated, the transcription factor
relocates to the nucleus where it activates its target genes.
Generally the sensor and TF are unique for a given stress
response pathway unlike transducers which can be shared
between different stress response pathways, leading to
what is commonly referred to as ‘crosstalk’ between the
pathways. A schematic diagram showing the general archi-
tecture of a stress response pathway is shown in Figure 1.
Oxidative stress response pathways
Oxidative stress is caused by exposure to reactive oxygen
intermediaries/species (ROS). The stress induced on the
cells by electrophiles and oxidants gives rise to a variety of
chronic diseases. The outcome of interactions between the
cell and oxidants is determined largely by the balance
between the enzymes that activate the reactive intermedi-
aries and the enzymes that detoxify these reactive interme-
diaries [8]. For example, oxidative stress contributes to
aging and age-related diseases such as cancer, cardiovascu-
lar disease, chronic inflammation, and neurodegenerative
disorders. The body has developed a variety of counterac-
tive measures for combating oxidative stress. At elevated
concentrations of electrophiles the complex Keap1-Nrf2
(made up of the transcription factor Nrf 2 and sensor
Keap1) is broken and Nrf2 is liberated and transported
into the nucleus. Keap1 has been known to sequester Nrf2
in the cytoplasm and also leads to the proteasomal degra-
dation of Nrf2. Once the complex is broken, Nrf2 is phos-
phorylated and transported to the nucleus. Inside the
nucleus, Nrf2 forms heterodimers with small Maf proteins
(SMP) which then binds to the anti-oxidant response
element (ARE) and leads to the translation of antioxidant
genes, which produces Phase II detoxifying enzymes.
The purpose of this is to detoxify the electrophiles to
water soluble components. Thus in response to elevated
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 2 of 16
concentrations of electrophiles, various antioxidant pro-
teins are activated [9-13]. The schematic diagram for Nrf2
activation is shown in Figure 2. In the rest of this paper,
the term ARE will be interchangeably used to represent
either the antioxidant response element cis enhancer
sequence that is upstream of the gene promoters for the
antioxidant proteins or the antioxidant genes/proteins
themselves. The context will make it clear whether we are
referring to the regulatory sequence or to the resulting
gene/protein.
We next focus on the procedure by which Nrf 2 is
deactivated. This is carried out by other proteins that
stop translation of the antioxidant genes once the elec-
trophiles have been neutralized. For instance, the
Bach1-SMP complex has been known to bind to the
same region on the ARE as the Nrf 2-SMP complex.
Similarly, small Maf proteins are known to form homo-
dimers or heterodimers with other small Maf proteins.
These protein complexes are known to bind to the same
location on the ARE as the Nrf2-SMP complex. So, once
the electrophiles have been eliminated, these protein
complexes bind to the ARE and displace Nrf 2 which
is then transported back to the cytoplasm. In the
cytoplasm, it binds with Keap1, which directs its
Figure 1 General scheme of stress response pathways. This figure explains the general flow of information in stress response pathways.
Figure 2 Nrf2 Activation. Explains how Nrf2 is activated and how it is able to neutralize the free radicals.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 3 of 16
proteosomal degradation [14-17]. The schematic dia-
gram for Nrf2 deactivation is shown in Figure 3.
One of the byproducts of normal metabolism is the
production of a large number of free radicals. Oxidative
stress is caused by the production of free radicals in quanti-
ties beyond those that can be handled by the cellular
antioxidant system. Indeed, oxidative stress has been impli-
cated in the development of many age-related diseases,
including neurodegenerative ones, such as Alzheimer’s and
Parkinson’s, and in aging itself. In addition, excess free
radicals react with the nucleotides in the DNA resulting
in mutations in the long run. Although there are cellular
mechanisms to sense and repair the oxidative DNA
damage, mutations can accumulate over a period of time
and result in a major disease like cancer. In the next
section, we develop a Boolean network model for oxidative
stress response pathways. This network will be later utilized
to analyze different failure modes that can supress apopto-
sis and possibly lead to cancer.
Boolean network modeling of oxidative stress response
pathways
Before proceeding to the actual modeling of the specific
oxidative stress response pathways, we first formally
define the general terms ‘pathway’ and ‘Boolean Network’
following the detailed development in [5]. Given two
genes/proteins A and B and binary values a, b Î {0, 1},
we define the term pathway segment A
t:a,b
→B to mean that
if gene/protein A assumes the value a then gene/protein
B transitions to b in no more than t subsequent time
steps. A pathway is defined to be a sequence of pathway
segments of the form A
t1:a,b
→B
t2:b,c
→C.
A Boolean Network (BN), ϒ = (V, F ), on n genes is
defined by a set of nodes/genes V = {x1, ..., xn}, xi Î {0, 1},
i = 1, ..., n, and a list F = (f1, ..., fn), of Boolean functions,
fi: {0, 1}n ® {0, 1}, i = 1, ..., n [18]. The expression of each
gene is quantized to two levels, and each node xi repre-
sents the state/expression of the gene i, where xi = 0
means that gene i is OFF and xi = 1 means that gene i is
Figure 3 Nrf2 Deactivation. Explains how after neutralizing free radicals Nrf2 is transported back to cytoplasm from mitochondria.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 4 of 16
ON. The function fi is called the predictor function for
gene i. Updating the states of all genes in ϒ is done syn-
chronously at every time step according to their predictor
functions. At time t, the network state is given by x(t) =
(x1(t), x2 (t), ..., xn(t)), which is also called the gene activity
profile (GAP) of the network.
The modeling approach that we will follow here
involves using the biological pathway knowledge from
the literature and applying Karnaugh map reduction
techniques to it to obtain an update equation for each
node of the Boolean network [5]. The details specific to
the oxidative stress response pathway are discussed
next. The pathway segments relevant to the oxidative
stress response are given below [9,10,12,15,19,20]:
ROS
1:1,0
→K eap1
(1)
ROS
1:1,1
→PKC
(2)
ROS
1:a,¯a
→Bach 1
(3)
Keap 1
1:b,¯b
→Nrf2
(4)
Nrf2, ROS
1:(1,0),1
→
Keap1
(5)
PKC
1:1,1
→Nrf2
(6)
Bach1, SMP
1:(1,1),0
→
ARE
(7)
Nrf2, SMP
1:(1,1),1
→
ARE
(8)
SMP, SMP
1:(1,1),0
→
ARE
(9)
ARE
1:1,1
→SMP
(10)
ARE
1:1,0
→ROS
(11)
ARE
1:1,0
→PKC
(12)
In these pathways ARE represents the family of antioxi-
dant genes in the sense that if the correct complexes bind
to ARE it leads to the up-regulation/down-regulation of
the appropriate antioxidant gene. These pathway interac-
tions are pictorially represented in Figure 4. In this figure
we have used square boxes without making any distinction
between whether they represent proteins/genes or a bio-
chemical entity. ROS stands for reactive oxidative species
which is a biochemical entity. The other entities like PKC,
Keap1, Nrf2, Bach1 are all proteins and ARE (Antioxidant
Response Element) is a cis enhancer sequence that is
upstream of the gene promoters for the antioxidant pro-
teins or the antioxidant genes/proteins themselves. Also
the merged activation (Nrf2/SMP) or inhibition (Bach1/
SMP) corresponds to dimers formed between these com-
ponents. The Karnaugh-maps for the genes/proteins are
shown in Figure 5.
From the pathways described above and using the
Karnaugh-map reduction techniques, the Boolean update
equations for each node of the network are deduced.
Some logical reasoning has been used for determining the
equations: 1) the maximum number of predictors for
updating a variable is fixed to be 3; 2) Small Maf Protein is
assumed to be ubiquitously expressed and the pathway
given by Eqn.(10) only increases the concentration of
SMP, which in conjunction with Eqn.(9), binds to ARE
and down-regulates the antioxidant gene; 3) a gene being
turned on implies that the corresponding protein is being
produced although, in reality, this is not necessarily true;
and 4) in the case of a conflict in the Karnaugh map, bio-
logical knowledge has been used to assign either a 0 or a
1. This last point is demonstrated by a specific example.
For instance, in the case of ARE, the entry shown with a
grey circle around it says that when both Bach1 and Nrf2
are upregulated and antioxidant gene is downregulated,
then at the next time step antioxidant gene will be upregu-
lated. The biological explanation for such an update is that
it corresponds to the situation where, in the presence of
Stress, Nrf2 has been activated and is relocating to the
nucleus while the inhibitor Bach1 is simultaneously relo-
cating to the cytoplasm prior to the activation of antioxi-
dant gene at the next time step. Such intuitive reasoning
has been used to model the system here. One might use a
different reasoning which could lead to a different set of
update equations. However, since we are concerned only
about the final steady-state behavior, such reasoning can
be justified as long as the overall system behavior, defined
by the update equations, matches the steady-state. As an
example, the final update equation for ARE is derived as
follows. In the K-maps, the ones are grouped up in pairs
of 2,4,8 and so on and each group should have at least one
variable staying constant. So for this case there are two
groups whose equations correspond to Nrf2 · (ARE) and
Nrf2 · (Bach1). The final update equation for ARE is the
sum of these two equations. Please refer to Additional file
1 for some additional details. Indeed, by working with dif-
ferent sets of update equations, we determined that all bio-
logically plausible ones led to the same/similar attractor
behavior. From the set of possible Boolean networks we
chose the ones that appealed most to our biological under-
standing and the resulting update equations are given
below:
ROSnext = Stress · ARE
(13)
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 5 of 16
Keap1next = ROS · (Nrf2 + Keap1)
(14)
PKCnext = ROS · ARE
(15)
Nrf2next = PKC + Keap1
(16)
Bach1next = ROS
(17)
Figure 4 Oxidative Stress Response Pathways. The major pathways involved in oxidative stress response.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 6 of 16
SMPnext = 1
(18)
AREnext = Nrf2 · (ARE + Bach1).
(19)
An equivalent digital circuit with logic gates is shown
in Figure 6. Here the lines in bold represent feedback
paths. The state transition diagrams resulting from Eqns.
(13)-(19) for the two cases Stress = 0 and Stress = 1 are
shown in Figures 7 and 8 respectively. In these transition
diagrams, the genes in the binary state representation are
ordered as [ROS Keap1 PKC Nrf2 Bach1 ARE] and the
binary states are compactly represented by their decimal
equivalents. For instance, the binary state (111100) would
be represented by the decimal number 60. The states of
particular interest are the attractors as they give rise to
the steady-state properties of the network. In Figure 7,
the state of interest is the singleton attractor 18(010010).
On the other hand, in Figure 8, the states of interest are
the seven states forming the attractor cycle. These states
are: 18(010010), 50(110010), 40(101000), 44(101100),
45(001101), 5(000101) and 23(010111) traversed in that
order. They would lead to cyclical/oscillatory behavior in
the time domain response.
It is clear from the preceding discussion that some kind
of oscillatory behavior of the genes will be observed when
the external Stress input equals 1. On the other hand,
when the Stress input equals 0, the system will rest in only
one state meaning that there will be no oscillation.
Time domain simulation results
The network obtained was simulated using MATLAB by
giving an external stress input signal for a duration of 50
timesteps, and both the input signal and the responses are
shown in Figure 9. The signal ROS is a biological manifes-
tation of the external input signal, Stress being applied to
the network. The biological purpose of this network is to
counteract the effect of ROS produced in response to the
Stress input. As we can see from Figure 9, in the absence
Figure 5 Karnaugh Maps for Deriving the Oxidative Stress Response Boolean Network. K-map simplification for all the elements involved
in the system.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 7 of 16
Figure 6 Equivalent Boolean Network for Oxidative Stress Response. Boolean network model for oxidative stress response based on the
equations derived using K-maps.
Figure 7 The Boolean State Transition Diagram when the Stress input is 0. The state transition diagram for the Boolean network with no
stress on the system. This gives us an idea of the attractor states of the system.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 8 of 16
of any Stress signal, the system reaches the singleton
attractor 18(010010). Once Stress signals are applied, there
are oscillations as theoretically expected from the exis-
tence of an attractor cycle. In Reichard et al. [14], the cells
were treated with Arsenite, a well known activator of Nrf2
and an out-of-phase relationship was observed between
Nrf2 and Bach1. Shan et al. [17] also showed a similar out
of phase relationship. In Katsuoka et al. [16]DEM (an
Figure 8 The Boolean State Transition Diagram when the Stress input is 1. The state transition diagram for the Boolean network with
stress on the system. This gives us an idea of the attractor states of the system.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 9 of 16
activator of Nrf2) also leads to increased expression of
NQO1 which is a known anti-oxidant response element.
Such an in-phase relationship between Nrf2 and the anti-
oxidant gene is also seen in Figure 9. Thus the theoretical
predictions made by our Boolean network model for oxi-
dative stress response appear to be consistent with experi-
mental observations from the published literature. Note,
however, that these experiments consider only two genes/
proteins at a time and therefore, there is a need for experi-
mentally studying the simultaneous activities of ROS,
Keap1, Nrf2, PKC, Bach1 and ARE in the time domain.
Mitochondria and free radical generation
Mitochondria play an important role in cellular energy
metabolism, free radical generation and apoptosis. It has
long been suspected that mitochondrial functions con-
tribute to the development and progression of cancer
[21-23]. Over 70 years ago, Otto Warburg proposed that
a key event in carcinogenesis is a defect in the respira-
tory mechanism, leading to increased glycolysis even in
the presence of oxygen;this is known as the Warburg
effect [24]. The well known function of mitochondria is
to generate Adenosine Triphosphate (ATP) molecules
providing energy for the survival of the cell through oxi-
dative phosphorylation (OXPHOS), which is collectively
accomplished by proteins encoded both by nuclear and
mitochondrial DNA. Oxidative phosphorylation is a
metabolic process, which takes place in mitochondria in
which ATP is formed as a result of the transfer of elec-
trons from NADH or FADH2 to O2 by a series of elec-
tron carriers. OXPHOS is the major source of ATP as
well as free radical generation in aerobic organisms. For
example, oxidative phosphorylation generates 26 of the
30 molecules of ATP that are formed when a molecule
of glucose is completely oxidized to CO2 and H2O,
although 1 to 2% of the electrons are lost during trans-
fer through the chains leading to free radical generation
[25]. Figure 10 [26] shows a schematic representation of
the whole process along with free radical generation.
The points shown with red stars correspond to the loca-
tions where free radicals are generated.
Even though it has been long recognized that increased
ROS production in mitochondria leads to genetic instabil-
ity and progression of cancer, there remain several unan-
swered questions regarding the complex signalling
capacity of this organelle [27]. The DNA is highly suscep-
tible to free radical attacks. Free radicals can break DNA
strands or delete bases. These mutations can prove to be
carcinogenic. It has been estimated that more than 10,000
hits of oxidative stress occur each day. So it is important
to tackle these free radicals at the source of their genera-
tion, which is why the mitochondria is also a very rich
source of anti-oxidants. Although cellular mechanisms can
tackle this stress, damage accumulates with age. At present
Figure 9 Time response behaviour of the system in Fig.4. Time response simulation of the Boolean network to observe oscillations of the
proteins in the system.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 10 of 16
altered energy metabolism is considered to be an addi-
tional hall mark of cancer progression [28] and these
metabolic pathways have been investigated as targets for
cancer therapy. In this paper, we will specifically focus on
the PI3k/Akt pathway which is one such pathway and is
described in the following section.
An integrated network for oxidative stress response
and apoptosis
Cancer is an umbrella term for diseases that are associated
with loss of cell-cycle control, leading to uncontrolled cell
proliferation and/or reduced apoptosis. It is often caused
by genetic alterations leading to malfunctioning in the bio-
logical pathways [1,29,30]. One of the possible cellular
responses resulting from oxidative stress is the induction
of apoptosis. Thus it is important to develop a network
model linking the oxidative stress input to the fate of the
cell. In this section, we will do precisely that by consider-
ing the oxidative stress response pathways alongside other
downstream pathways capable of inducing apoptosis. Spe-
cifically, we will focus on the PI3k/Akt pathway. The PI3k/
Akt pathway is downstream of the Ras gene which is
known to play an important role in many cancers. In addi-
tion, other genes in the PI3k/Akt pathways are found
mutated in many cases of cancer. Oxidative stress often
upregulates many of the genes in the PI3k/Akt pathway.
The detailed interactions between the oxidative stress
response pathway and the PI3k/Akt pathway are shown in
Figure 11 [1,31-33]. Starting with this pathway diagram
and utilizing the procedure developed earlier in Section,
an equivalent digital circuit in terms of logic gates can be
implemented as shown in Figure 12. The above circuit is
modeled with two output genes which effectively control
the final fate of the cell. Bad and Bcl2 are known to have
pro-apoptotic and anti-apoptotic functions respectively
and thus can serve as biomarkers of apoptosis induction.
Indeed, it is the delicate balance between the activity of
these two genes that dictates the ultimate fate of the cell
[34-36]. The purpose of the Nrf 2-ARE pathway in this
integrated network is to reduce the average value of ROS
present in the system, in response to the oxidative stress.
This is clear from the plot in Figure 9: between the time
instants from the 25th timestep to the 75th timestep when
there is a continuous Stress present in the system, the ROS
present in the system is oscillating between 0 and 1 which
implies that its average value is less than ‘1’, which is the
value that we would have otherwise had in the absence of
the Nrf 2-ARE pathway.
Classification of faults in the integrated network
In the integrated pathway diagram of Figure 11, the two
genes namely Bad and Bcl2 are instrumental in deciding
the fate of the cell. The preferred status of the two
genes, when oxidative stress is not being neutralized, are
1 and 0 respectively since it corresponds to the situation
where the pro-apoptotic factor is turned ON and the
anti-apoptotic factor is turned OFF. Although a devia-
tion from this state may not signal that the cell is turn-
ing cancerous, there is a higher possiblity of the cell
exhibiting aberrant behaviour.
Depending on the final resting status of these two genes,
one may be able to characterize the degree of invasiveness
of the disease especially if it is being caused by apoptosis
supression. Once it has been determined that a cell is
Figure 10 Stages of Oxidative Phosphorylation producing free radicals. Explains Krebs cycle and how and where free radicals are produced
in the mitochondria.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 11 of 16
exhibiting abberant behavior, one would like to pinpoint
the location of the fault/error so that the necessary thera-
peutic intervention(s) can be applied. Since the digital cir-
cuit model of Figure 12 uses logic gates, it should be
possible to use the fault detection techniques from the
Digital Logic literature [37,38] to pinpoint the fault loca-
tions. This will be carried out in this section. An important
difference between the results obtained in Layek et al. [6]
for pinpointing the fault locations in the MAPKinase path-
ways and the results to be presented here is that the digital
circuit in Figure 12 involves feedback and its behaviour is,
therefore, much more complicated to analyze. However, it
should be pointed out that the simpler fault pinpointing
methodology presented in Layek et al. [6] is much more
amenable to biological validation via appropriately
designed experiments while the same cannot be said about
the results to be presented here. Indeed, the results to be
presented here show that the pinpointing of the fault loca-
tions is theoretically possible even in this case, although
the biological feasibility of the methods required is open
to question.
We note that the faults in a digital circuit are mainly
of two types [37]:
• Stuck-at Faults: As the name implies, this is a fault
where a particular line l is stuck at a particular value
a Î {0, 1}, denoted by line l,s-a-a (s-a-a means
stuck-at-a). This means that the value at that line is
always going to be a regardless of the inputs coming
in. This can be thought of as something similar to a
mutation in a gene, where a particular gene is either
permanently turned ON or OFF.
• Bridging Faults: This is the type of fault where new
interconnections are introduced among elements of
the network. This can be thought of as new pathways
being created in the cell. This type of fault is not con-
sidered in the current paper due to the lack of biologi-
cal knowledge about new pathways being introduced.
Here, it is appropriate to mention that the biological
relevance of each of these two types of faults has been
discussed in Layek et al. [6].
Figure 11 Pathway Diagram of Oxidative Stress along with PI3k/Akt. Inclusion of PI3k/Akt pathways along with oxidative stress pathways
and study how they can lead to aberrant be-haviour in cells.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 12 of 16
The digital circuit in Figure 12 has feedback (shown in
bold lines) and is, therefore, a sequential circuit. To
detect a fault in a sequential circuit we need a test
sequence. Let T be a test sequence and let R(q,T) be the
response of the fault-free sequential system N starting in
the intial state q. Now let the faulty sequential circuit be
denoted by Nf where f is the fault. Let us denote by Rf
(qf,T) the response of Nf to T starting in the initial state
qf. A test sequence T detects a fault f iff (if and only if
or equivalently this condition is both necessary and suf-
ficient) for every possible pair of initial states q and qf,
the output sequences R(q,T) and Rf (qf ,T) are different
for some specified vector ti Î T. The output being
observed is the status of [Bad, Bcl2].
Once this output shows a deviation from a desired value,
it becomes imperative to pinpoint the possible fault loca-
tions which can give rise to the aberrant behaviour. To do
so, one can represent the digital circuit of Figure 12 as in
Figure 13. The Primary Input(PI) is Stress which is the
only external signal which the experimenter has control
over. The Primary Output’s (PO’s) are the status of Bad
and Bcl2, which are the only outputs available to the
experimenter. The Secondary Output’s and Secondary
Input’s are [ARE, Keap1, Mdm2], which are being fed back
into the system. The states of these 3 genes ARE, Keap1
and Mdm2 determine the internal state of the system.
These 3 elements can be considered as memory elements
of the system as their previous state is retained by the
system and fed back. The input sequence consists of two
parts namely a Homing sequence and a Test sequence,
denoted by H and T respectively.
The purpose of this procedure is to pinpoint the possi-
ble locations for the fault f in Nf, given the output
sequence of Bad and Bcl2 for the normal and faulty cir-
cuits. It is assumed that we have no knowledge about the
initial status of any of the genes. Knowledge of the initial
status of the internal states is important as all future
computations are based on these values. The Homing
sequence is an initial input sequence that brings the net-
work to a known internal state. So, once the Homing
Sequence is given to N and Nf, N will come to a known
internal state. Note that a similar claim cannot be made
about Nf as the fault f is not known apriori. For the cir-
cuit in Figure 13, a possible Homing sequence is [0 0 0 0
0 0 0 0], which brings the internal state of the system to
[0 1 0]. This means that if the Stress input is zero for
eight time steps, then at the end of that period, the inter-
nal state of the system becomes [0 1 0], regardless of the
initial status of any of the genes in the network. A reason
for choosing this Homing sequence is that it implies that
no input needs to be given to the system and it evolves to
the indicated internal state. In future when we are trying
to validate these results experimentally this will be of
immense help. If we refer back to Figure 7, we see that
regardless of the initial state, within four time steps the
trajectory reaches the state (’010010’) where ARE = 0 and
Figure 12 Boolean Network modeling of Fig. 11. A Boolean network model of the network along with PI3k/Akt pathways.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 13 of 16
Keap1 = 1. This is consistent with the conclusion that we
are getting from the Homing sequence here with the only
difference that a slightly longer sequence is required here
as the state transition diagram has a higher cardinality
than that in Figure 7.
Once the Homing sequence has done its job, the Test
sequence(T) is fed into N and Nf, and by comparing the
output states of the normal and faulty networks, we can
pinpoint the location of the fault in the network, assuming
that a single stuck-at-fault has occurred. This can be car-
ried out using the time-frame expansion method which is
briefly discussed next. The block in Figure 13 is replicated
n times with the feedback loops cut-off. The Secondary
Output of the kth stage is fed as the Secondary Input for
the (k + 1)th stage. The Primary Outputs of the first (n - 1)
stages are neglected. The Primary Outputs of the nth stage
of the normal and faulty circuits will be different as the
network configurations are different for both. The Primary
Input sequence has to be derived so that the error in a line
is propagated to the primary output in n time steps, so
that a difference is observed at the primary outputs of the
normal and faulty circuits [37,38]. The situation is picto-
rially represented in Figure 14. Please refer to Additional
file 1 for to y example.
From the preceding discussion in this section, we know
that there are 15 possible genes (this is the total number
of genes in Figure 11, excluding the output genes Bad
and Bcl2) where there could be a mutation. This means
that there are 30 cases of faults as a single gene can be
mutated as a s-a-0 or as a s-a-1. We consider all possible
cases of single mutation, because in the presence of
mutation, the normal and faulty system cannot produce
the same output unless, of course, the mutated gene is
not a critical one. Based on the methods described earlier
in this section, we came up with a list of test sequences
for the detection of each gene fault. It is to be noted that
the Test Sequences generated here are only for the Hom-
ing Sequence considered earlier. For a different Homing
Sequence the Test Sequence will also be different. The
different test sequences and their ability to detect differ-
ent single stuck-at faults are tabulated in Figure 15. Here,
truncated versions of the same test sequence can be used
to detect different faults appearing in the same row. For
detecting any particular fault, one would apply the test
sequence from the same row truncated at the bit whose
color matches that of the particular fault. The mismatch
between the outputs of the normal and faulty systems,
characterized by the vector [Bad, Bcl2] would then result
in the detection of that fault. Thus we have developed a
method to pinpoint the possible fault locations in a Boo-
lean network with feedback. The algorithm will work
with multiple fault cases too with minor modifications.
Concluding remarks
In this paper, we have developed a Boolean network
model for the oxidative stress response. This model was
developed based on pathway information from the cur-
rent literature pertaining to oxidative stress. Where
applicable, the behaviour predicted by the model is in
agreement with experimental observations from the
published literature. It is our hope that some of the
additional predictions here, such as those pertaining to
Figure 13 Block Diagram Representation of Fig.12. A simple description of the system showing clearly the feedback lines in the system.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 14 of 16
the oscillatory behaviour of certain genes in the pre-
sence of oxidative stress, will be experimentally validated
in the near future.
We have also linked the oxidative stress response to
the phenomenon of apoptosis via the PI3k/Akt pathway.
An integrated model based on collectively considering
the PI3k/Akt pathways and the oxidative stress response
pathways was developed and then used to pinpoint pos-
sible fault locations based on the Bad-Bcl2 apoptotic
signatures in response to ‘test’ oxidative stress inputs.
The approach used to achieve this differs significantly
from the earlier results in Layek et al. [6] since the Boo-
lean network of this paper has feedback. The approaches
used here and in Layek et al. [6] could potentially have
a significant effect on cancer therapy in the future as
pinpointing the possible fault location(s) in cancer could
permit the choice of the appropriate combination of
drugs (such as kinase inhibitors) for maximum thera-
peutic effectiveness. Of course, it should be pointed out
that the theoretical procedure presented here for pin-
pointing fault locations in a biological network with
feedback will need to be further simplified before it can
be even considered for practical biological validation.
Additional material
Additional file 1: Explains the algorithms discussed in the
manuscript with toy examples.
Acknowledgements
Based on “Modelling oxidative stress response pathways”, by Sriram
Sridharan, Ritwik Layek, Aniruddha Datta and Jijayanagaram Venkatraj which
appeared in Genomic Signal Processing and Statistics (GENSIPS), 2011 IEEE
International Workshop on. © 2011 IEEE [39].
This work was supported in part by the National Science Foundation under
Grants ECCS-0701531 and and ECCS-1068628 and in part by the J. W.
Runyon, Jr. ‘35 Professorship II Endowment Funds at Texas A & M University.
This article has been published as part of BMC Genomics Volume 13
Supplement 6, 2012: Selected articles from the IEEE International Workshop
Figure 14 Fault Detection using Time-Frame Expansion. Fault detection in the boolean(digital) network using time-frame expansion method.
Figure 15 Test Sequences for detecting single stuck-at-faults. The test sequence which can be given to system to find out single stuck-at-
faults based on output signature.
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 15 of 16
on Genomic Signal Processing and Statistics (GENSIPS) 2011. The full
contents of the supplement are available online at http://www.
biomedcentral.com/bmcgenomics/supplements/13/S6.
Author details
1Texas A & M University, Electrical and Computer Engineering, College
Station, TX, 77843-3128, USA. 2Texas A & M University, Vet Integrative
Biosciences, College Station, TX, 77843-4458, USA.
Authors’ contributions
Sriram Sridharan did most of the theoretical work on this paper with some
assistance from Ritwik Layek. Aniruddha Datta provided overall direction and
supervision while Jijayanagaram Venkatraj provided the relevant supporting
biological domain knowledge.
Competing interests
The authors declare that they have no competing interests.
Published: 26 October 2012
References
1.
Weinberg RA: The Biology of Cancer. Garland Science, Princeton;, 1 2006.
2.
Datta A, Dougherty E: Introduction to Genomic Signal Processing with Control
Boca Raton: CRC Press; 2007.
3.
Viswanathan GA, Seto J, Patil S, Nudelman G, Sealfon SC: Getting Started in
Biological Pathway Construction and Analysis. PLoS Comput Biol 2008, 4:
e16.
4.
Saraiya P, North C, Duca K: Visualizing biological pathways: requirements
analysis, systems evaluation and research agenda. Information
Visualization 2005, 1-15.
5.
Layek R, Datta A, Dougherty ER: From Biological Pathways to Regulatory
Networks. Mol BioSyst 2011, 7:843-851.
6.
Layek R, Datta A, Bittner M, Dougherty ER: Cancer therapy design based
on pathway logic. Bioinformatics 2011, 27:548-555.
7.
Simmons SO, Fan CY, Ramabhadran R: Cellular Stress Response Pathway
System as a Sentinel Ensemble in Toxicological Screening. Toxicol Sci
2009, 111:202-225.
8.
Storz G, Imlay JA: Oxidative Stress. Current Opinion in Microbiology 1999,
2:188-194.
9.
Kensler TW, Wakabayashi N, Biswa S: Cell Survival Responses to
Environmental Stresses Via the Keap1-Nrf2-ARE Pathway. Annu Rev
Pharmacol Toxicol 2007, 47:89-110.
10.
Lee JM, Johnson JA: An Important Role of Nrf2-ARE Pathway in the
Cellular Defense Mechanism. J Biochem Mol Biol 2004, 37:139-143.
11.
Hong F, Freeman KRSML, Liebler DC: Specific patterns of electrophile
adduction trigger Keap1 ubiquitination and Nrf2 activation. J Biol Chem
2005, 280:31768-75.
12.
Itoh K, Wakabayashi N, Katoh Y, Ishii T, O’Connor T, Yamamoto M: Keap1
regulates both cytoplasmic-nuclear shuttling and degradation of Nrf2 in
response to electrophiles. Genes to Cells 2003, 8:379-391.
13.
Niture SK, Jaiswal AK: Prothymosin Mediates Nuclear Import of the INrf2/
Cul3_Rbx1 Complex to Degrade Nuclear Nrf2. The Journal of Biological
Chemistry 2009, 284:13856-68.
14.
Reichard JF, Motz GT, Puga A: Heme oxygenase-1 induction by NRF2
requires inactivation of the transcriptional repres-sor BACH1. Nucl Acids
Res 2007, 35:7074-86.
15.
Dhakshinamoorthy S, Jain AK, Bloom DA, Jaiswal AK: Bach1 competes with
Nrf2 leading to negative regulation of the antioxidant response element
(ARE)-mediated NAD(P)H:quinone oxidoreductase 1 gene expression and
induction in response to antioxidants. J Biol Chem 2005, 280:16891-900.
16.
Katsuoka F, Motohashi H, Engel JD, Yamamoto M: Nrf2 Transcriptionally
Activates the mafG Gene through an Antioxidant Response Element.
J Biol Chem 2005, 280:4483-90.
17.
Shan Y, Lambrecht RW, Donohue SE, Bonkovsky HL: Role of Bach1 and
Nrf2 in up-regulation of the heme oxygenase-1 gene by cobalt
protoporphyrin. FASEB J 2006, 20:2651-3.
18.
Kauffman S: The Origins of Order: Self-Organization and Selection in Evolution
New York: Oxford Univ. Press; 1993.
19.
Lee OH, Jain AK, Papusha V, Jaiswal AK: An Auto-regulatory Loop between
Stress Sensors INrf2 and Nrf2 Controls Their Cellular Abundance. The
Journal of Biological Chemistry 2007, 282:36412-36420.
20.
Huang HC, Nguyen T, Pickett CB: Phosphorylation of Nrf2 at Ser-40 by
Protein Kinase C Regulates Antioxidant Response Element-mediated
Transcription. The Journal of Biological Chemistry 2002, 277:42769-42774.
21.
Goh J, Enns L, Fatemie S, Hopkins H, Morton J, Pettan-Brewer C, Ladiges W:
Mitochondrial targeted catalase suppresses invasive breast cancer in
mice. BMC Cancer 2011, 11:191.
22.
Sotgia F, Martinez-Outschoorn UE, Lisanti MP: Mitochondrial oxidative
stress drives tumor progression and metastasis: should we use
antioxidants as a key component of cancer treatment and prevention.
BMC Medicine 2011, 9:62.
23.
Bai RK, Leal SM, Covarrubias D, Liu A, Wong LJC: Mitochondrial Genetic
Background Modifies Breast Cancer Risk. Cancer Res 2007, 67:4687-4694.
24.
Ferreira LM: Cancer metabolism: The Warburg effect today. Experimental
and Molecular Pathology 2010, 89:372-380.
25.
Inoue M, Sato EF, Nishikawa M, Park Ah-Mee, Kira Y, Imada I, Utsumi K:
Mitochondrial Generation of Reactive Oxygen Species and its Role in
Aerobic Life. Current Medicinal Chemistry 2003, 10:2495-2505.
26.
Beal MF: Less stress, longer life. Nature Medicine 2005, 11:598-599.
27.
Sitaramayya A: Signal Transduction: Pathways, Mechanisms and Diseases
Berlin: Springer Verilag; 2010.
28.
Hanahan D, Weinberg RA: Hallmarks of Cancer: The Next Generation. Cell
2011, 144:646-674.
29.
Hanahan D, Weinberg RA: The Hallmarks of Cancer. Cell 2000, 100:57-70.
30.
Hahn WC, Weinberg RA: Rules for Making Human Tumor Cells. N Engl J
Med 2002, 1593-1603.
31.
Sakamoto K, Iwasaki K, Sugiyama H, Tsuji Y: Role of the Tumor Suppressor
PTEN in Antioxidant Responsive Element-mediated Transcription and
Associated Histone Modifications. Mol Biol Cell 2009, 20:1606-17.
32.
Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB: Exploiting the PI3K/AKT
Pathway For Cancer Drug Discovery. Nature Reviews, Drug Discovery, 2005,
4:988-1004.
33.
Wang L, Chen Y, Sternberg P, Cai J: Essential Roles of the PI3 Kinase/Akt
Pathway in Regulating Nrf2-Dependent Antioxidant Functions in the
RPE. Invest Ophthalmol Vis Sci 2008, 49:1671-1678.
34.
Martindale JL, Holbrook NJ: Cellular Response to Oxidative Stress:
Signaling for Suicide and Survival. Journal of Cellular Physiology 2002,
192:1-15.
35.
Gross A, McDonnell JM, Korsmeyer SJ: BCL-2 family members and the
mitochondria in apoptosis. Genes and Development 1999, 13:1899-1911.
36.
Coultas L, Strasser A: The role of the Bcl-2 protein family in cancer.
Seminars in Cancer Biology 2003, 13:115-123.
37.
Abramovici M, Breuer MA, Friedman AD: Digital Systems Testing and
Testable Design. IEEE Press, The IEEE, Inc, New York;, 1 1990.
38.
Breuer MA, Friedman AD: Diagnosis and Reliable Design of Digital
Systems. Computer Science Press, Inc., Maryland; 1989.
39.
Sridharan S, Layek R, Datta A, Venkatraj J: Modelling oxidative stress
response pathways. Genomic Signal Processing and Statistics (GENSIPS), 2011
IEEE International Workshop on: 4-6 December 2011 2011, 166-169.
doi:10.1186/1471-2164-13-S6-S4
Cite this article as: Sridharan et al.: Boolean modeling and fault
diagnosis in oxidative stress response. BMC Genomics 2012 13(Suppl 6):
S4.
Submit your next manuscript to BioMed Central
and take full advantage of:
• Convenient online submission
• Thorough peer review
• No space constraints or color figure charges
• Immediate publication on acceptance
• Inclusion in PubMed, CAS, Scopus and Google Scholar
• Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Sridharan et al. BMC Genomics 2012, 13(Suppl 6):S4
http://www.biomedcentral.com/1471-2164/13/S6/S4
Page 16 of 16
|
23134720
|
ARE = ( ( ( ( Nrf2 ) AND NOT ( GSK3b ) ) AND NOT ( Bach1 ) ) AND NOT ( ARE ) )
Ras = ( ROS )
PTEN = NOT ( ( ROS ) )
Akt = ( PIP3 )
p53 = ( ( ATM ) AND NOT ( Mdm2 ) )
Bad = NOT ( ( Akt ) )
PI3K = ( Ras )
Nrf2 = ( ( Akt ) OR ( PKC ) ) OR NOT ( Akt OR Keap1 OR PKC )
Keap1 = ( ( Keap1 ) AND NOT ( Bach1 ) ) OR ( ( Nrf2 ) AND NOT ( Bach1 ) )
PIP3 = ( ( PIP2 ) AND NOT ( PTEN ) )
Mdm2 = ( ( Akt ) AND NOT ( ATM ) ) OR ( ( p53 ) AND NOT ( ATM ) )
ROS = ( ( Stress ) AND NOT ( ARE ) )
GSK3b = NOT ( ( Akt ) )
Bach1 = NOT ( ( ROS ) )
PIP2 = ( PI3K )
PKC = ( ROS AND ( ( ( NOT ARE ) ) OR ( ( NOT ARE ) ) ) )
Bcl2 = NOT ( ( Bad ) OR ( p53 ) )
ATM = ( ROS )
|
ORIGINAL RESEARCH ARTICLE
published: 10 December 2012
doi: 10.3389/fphys.2012.00446
Boolean model of yeast apoptosis as a tool to study yeast
and human apoptotic regulations
Laleh Kazemzadeh1,2, Marija Cvijovic 1,3* and Dina Petranovic 1*
1 Department of Chemical and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
2 Digital Enterprise Research Institute, National University of Ireland, Galway, Ireland
3 Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
Edited by:
Matteo Barberis, Humboldt University
Berlin, Germany; Max Planck Institute
for Molecular Genetics, Berlin,
Germany
Reviewed by:
Ioannis Xenarios, SIB Swiss Institute
of Bioinformatics, Switzerland
Abhishek Garg, Harvard University,
USA
*Correspondence:
Marija Cvijovic, Department of
Mathematical Sciences, Chalmers
University of Technology and
University of Gothenburg, Chalmers
tvärgata 3, Göteborg 412 96, Sweden.
e-mail: marija.cvijovic@chalmers.se;
Dina Petranovic, Department of
Chemical and Biological Engineering,
Chalmers University of Technology,
Kemivägen 10, Göteborg SE-412 96,
Sweden.
e-mail: dina.petranovic@chalmers.se
Programmed cell death (PCD) is an essential cellular mechanism that is evolutionary con-
served, mediated through various pathways and acts by integrating different stimuli. Many
diseases such as neurodegenerative diseases and cancers are found to be caused by, or
associated with, regulations in the cell death pathways.Yeast Saccharomyces cerevisiae, is
a unicellular eukaryotic organism that shares with human cells components and pathways
of the PCD and is therefore used as a model organism. Boolean modeling is becoming
promising approach to capture qualitative behavior and describe essential properties of
such complex networks. Here we present large literature-based and to our knowledge first
Boolean model that combines pathways leading to apoptosis (a type of PCD) in yeast. Analy-
sis of the yeast model confirmed experimental findings of anti-apoptotic role of Bir1p and
pro-apoptotic role of Stm1p and revealed activation of the stress protein kinase Hog propos-
ing the maximal level of activation upon heat stress. In addition we extended the yeast
model and created an in silico humanized yeast in which human pro- and anti-apoptotic reg-
ulators Bcl-2 family andValosin-contain protein (VCP) are included in the model.We showed
that accumulation of Bax in silico humanized yeast shows apoptotic markers and that VCP
is essential target of Akt Signaling. The presented Boolean model provides comprehen-
sive description of yeast apoptosis network behavior. Extended model of humanized yeast
gives new insights of how complex human disease like neurodegeneration can initially be
tested.
Keywords: apoptosis, Boolean modeling, Stm1, Bir1, Hog1,VCP, Bcl-2 family
INTRODUCTION
Apoptosis is a complex process which is strictly under control of
several regulatory networks. Any kind of malfunctioning in these
controlling systems due to insufficient or excessive apoptosis sig-
nal can potentially lead to threatening diseases such as various
types of cancer and neurodegenerative disorders. Therefore keep-
ing this process tightly regulated is important for the cell. Even
though apoptosis is often studied in multicellular organisms, the
discovery of yeast apoptosis in 1997 (Madeo et al., 1997) attracted
the attention of the wide research community (Frohlich et al.,
2007; Owsianowski et al., 2008; Madeo et al., 2009; Carmona-
Gutierrez et al., 2010b). As in other multicellular organisms the
apoptosis in yeast is triggered by both internal and external sig-
nals. In yeast,the external signals can include acetic acid (Ludovico
et al., 2001, 2002), salts, metal ions, ethanol, osmotic stress, heat
stress (Madeo et al., 2009), lipids (Aerts et al., 2008; Low et al.,
2008; Garbarino et al., 2009), mating pheromone (Zhang et al.,
2006), different pharmacological molecules, and drugs (Almeida
et al., 2008). Internal signals can include, ammonium, NO, ROS
(that can be generated within the cell by mitochondria and the
ER, and also induced by H2O2 addition (Madeo et al., 1999)
and other factors), damage (proteins, lipids, nucleic acids) as a
consequence of aging and mutations (Mazzoni et al., 2005; Wein-
berger et al., 2005; Hauptmann et al., 2006), as well as expression
of heterologous proteins, such as human pro-apoptotic proteins
(Eisenberg et al., 2007). Many proteins residing in the cytoplasm,
nucleus,mitochondria,ER,peroxisomes,and lysosomes have been
identified as the regulators of apoptosis. For example, proteoly-
sis is one of the main steps that leads to execution of cell death
and, a yeast metacaspase Yca1p has been shown to be central
for most (but not all) cell death scenarios (Madeo et al., 2002,
2009). Besides degradation of proteins, degradation of nucleic
acids is also carried out during apoptotic death and one of the two
important caspase-independent mediators is Nuc1p (homolog
of endonuclease G; Buttner et al., 2007). The second is Aif1p
(apoptosis-inducing factor; Wissing et al., 2004) that is together
with Nuc1p released from the mitochondrion and translocated
to the nucleus during the initiation and execution of apopto-
sis. The regulation of apoptosis in the nucleus, is achieved via
pro-apoptotic factor Nma111p (nuclear mediator of apoptosis;
Fahrenkrog et al., 2004) a serine protease that cleaves an anti-
apoptotic factor (inhibitor of apoptosis, IAP) Bir1p, which is
the only known IAP in yeast, and its anti-apoptotic mechanisms
(known to beYCA1-independent) are not well characterized (Wal-
ter et al., 2006). To understand how large and complex network
of apoptosis process is regulated it should be studied as a whole
allowing identification of the properties essential for biological
function (Janes et al., 2005). To complement experimental studies
www.frontiersin.org
December 2012 | Volume 3 | Article 446 | 1
Research Topic: From structural to molecular systems biology: experimental and computational approaches to unravel mechanisms of kinase activity
regulation in cancer and neurodegeneration
Kazemzadeh et al.
Boolean model of yeast apoptosis
mathematical models are often use permitting systematic analysis
of the network components either individually or jointly (Wolken-
hauer,2002; Stelling,2004). In Boolean networks (BN) introduced
by Kauffman (1969) these assumptions are made based on activa-
tion/inhibition effects of one node on another downstream node.
The Boolean “on” state (or 1 state or “true” state) can be translate
to biological active state of specific species, while “off” state (or
0 state or “false” state) corresponds to inactive state. With sim-
ple logical rules (AND, OR, and NOT) it is possible to capture
system’s behavior in a discrete manner without being dependent
on experimental measurements such as molecular concentration
or kinetic rates. This type of model implementation is becom-
ing more common in biology (Handorf and Klipp, 2012) and
examples include various model organisms and processes, rang-
ing from cell cycle models of simple fission yeast (Davidich and
Bornholdt, 2008), to complex dynamic analysis of mammalian
cell cycle (Fauré et al., 2006), study of mammalian neurotrans-
mitter signaling pathway (Gupta et al., 2007), investigation of
irreversible mammalian apoptosis and stable surviving (Mai and
Liu, 2009), and model of apoptosis in human (Schlatter et al.,
2009). In each of these studies different extensions of Boolean
modeling is implemented giving clear indication of the grow-
ing application of logic based modeling in qualitative studies of
biological networks where there is not much quantitative data
available.
We describe here a model based on Boolean network approach
that consists of two parts: in the first part construction and
the evaluation of the yeast apoptosis Boolean model is intro-
duced and in the second part we propose the use of established
model for study of human apoptotic proteins in yeast. Simplic-
ity of BN allowed us to construct the model that integrates vast
amount of heterogeneous knowledge that currently exists for yeast
apoptosis. The major purpose of this study is to understand the
emergence of systems properties. Extensive analysis of the state
space in combination of different input signals generated series
of in silico experiments. Simulating knock out experiments we
were able to test the function of specific feedback structures in
apoptotic network. The results were compared with the exist-
ing experimental data and the model was used to explore several
hypotheses in order to better understand certain apoptotic mech-
anisms and to suggest new strategies for further experimental
studies.
RESULTS
NETWORK TOPOLOGY
The constructed yeast apoptosis network contains 73 species
and 115 reactions (Table 1). Six species do not have succes-
sor (Sink/Output) or predecessor (Source/Input; Table 2). In the
schematic diagram (Figure 1) species are represented as nodes and
reactions as edges. Species include: processes, proteins, and small
molecules (metabolites or signals) and Reactions include activa-
tion (green arrow) and inhibition (red arrow). Nine nodes depict
inputs to the model and are colored blue. Seventeen elements are
active in nucleus and are shown in gray, 12 mitochondria species
are shown in yellow, and 34 orange boxes represent species resid-
ing in cytoplasm. System has only one output node which is called
“Apoptosis” and it is in dark blue. Filled blue circles are used as
an “AND” gates between two or more reactions to indicate the
necessity of presence of two species for activation or inhibition of
a reaction (Figure 1).
Network is activated via input signals corresponding to the fol-
lowing species: Acetic Acid, Heat, H2O2, Adozelesin, Mg2+, Cu2+,
Salt (NaCl), mating, and osmotic stress.
MODEL PROPERTIES
Like many molecular mechanisms in living cells, apoptosis can be
approximated as an outcome of sequential regulation steps which
do not occur all at the same time. As a result, upon induction,
the state of the cell changes with passing time. In order to cap-
ture this feature, each reaction was assigned activation time scale
implicating occurrences of different scenarios in sequential order.
By introducing time delays to the logical function, it is possible
to describe dynamic behavior of the given process using logical
networks (Thomas and D’Ari, 1990).
For this study we used Cell Net Analyser (CNA; Klamt et al.,
2006) which provides the function to capture signal propagation
in a time series to get a snapshot of the network and discrim-
ination of signaling events. This approach has been successfully
used to model human apoptotic network revealing new feedback
loops (Schlatter et al., 2009). To describe dynamic behavior of
the yeast apoptosis five timescales t = [0, 2, 4, 5, 6] are assigned.
These timescales are constants and indicate at which timescale
each node gets activated or inhibited. Simulation of the network
at t = x comprises of all interactions with timescale t ≤x but not
interactions with t ≥x. This gives the possibility of separation of
different functional groups such as different signaling pathways
or feedback loops. It should be noted that specified timescales do
not refer to real time but are only indicators of sequential regu-
latory steps (difference between timescale t = 0 and t = 2 is equal
to difference between timescales t = 2 and t = 4 or t = 4 and t = 5
or between t = 5 and t = 6). Timescales are not assigned based
on speed of reactions (how fast or slow reactions are), they are
assigned based on the sequence of events. To allow flexibility in
our model and facilitate changes and insertions of new events
timescales 1 and 3 are reserved and not used in the current net-
work structure. First timescale t = 0 is assigned to genes which
are already present and constantly active in cell (27 reactions).
Values for all input arcs to stimuli species are set to 1 at second
timescale t = 2 (53 reactions). Further interactions are activated
at time t = 4 (17 reactions). Although yeast apoptosis does not
contain any feedback loops time point t = 5 is reserved for feed-
back loops which get activated in response to osmotic and heats
shock and will lead to apoptosis. Considering impact of feed-
back loops on system behavior it is reasonable to assign them a
separate timescale. Finally interaction occurring at the very end
are assigned timescale t = 6 (18 reactions). Details on each reac-
tion, their time points and species involved in reactions are in
Table 3.
The model simulated the induction of apoptosis both in the
independent mode (assessing each individual stimulus separately)
as well as in the additive mode, where the activation of all inputs
were set at the same time (Table 4).
Frontiers in Physiology | Systems Biology
December 2012 | Volume 3 | Article 446 | 2
Kazemzadeh et al.
Boolean model of yeast apoptosis
Table 1 | List of species in yeast Boolean model.
ID
Species
Type
Name description
1
ABNORMALTELOMERASE
Change
ABNORMALTELOMERASE
2
ACETIC ACID
Input
Chemical
3
ADENYLATECYCLASE
Enzyme
Lyase enzyme
4
ADOZELESIN
Input
Drug
5
AIF-MT
Protein
Apoptosis-inducing factor in mitochondria
6
AIF-NUC
Protein
Apoptosis-inducing factor in nucleus
7
APOPTOSIS
Output
Cell death
8
BIR1
Protein
Baculoviral IAP repeat-containing protein 1
9
CAMP
Protein
Cyclic adenosine monophosphate
10
CDC48
Protein
Cell division cycle
11
CDC6
Protein
Cell division cycle
12
CPR3
Protein
Cyclosporin-sensitive proline rotamase
13
CU2
Input
Ion
14
DesCYCLINCCDK8
Change
Destruction of cylinC/CDCk8
15
CYTC-CYT
Protein
CytochromC in cytososl
16
CYTC-MT
Protein
CytochromC in mitochondria
17
DNA-FRAG
Change
DNA fragmentation
18
DRE2/TAH18
Change
Dre2-TAH18 complex
19
EMC4
Protein
ER membrane protein complex
20
ESP1
Protein
Separase
21
FIS1
Protein
Mitochondrial FISsion
22
FYV10
Protein
Function required for yeast viability
23
H2B
Protein
Histon 2B
24
H2O2
Input
Hydroxide peroxide
25
HEAT
Input
Event
26
HK
–
House keeping function**
27
HOG1
Protein
High osmolarity glycerol response
28
HOG1-DEP
Protein
HOG1 dependent genes
29
HOS3
Protein
Hda one similar
30
KAP123
Protein
KAryoPherin
31
MAPK
Protein
Map kinase cascade
32
MATING
Input
Mating pheromone
33
MCD1-MT
Protein
Mitotic chromosome determinant in mitochondria
34
MCD1-NUC
Protein
Mitotic chromosome determinant in nucleus
35
MDV1
Protein
Mitochondrial DiVision
36
MEC1
Protein
Mitosis entry checkpoint
37
MG2
Input
Ion
38
MMI1
Protein
Translation machinery associated
39
MSN2-4
Protein
Multicopy suppressor of SNF1 mutation
40
MT-ALT
Change
Mitochondria alteration
41
MT-FRAG
Change
Mitochondria fragmentation
42
NDI1
Protein
NADH dehydrogenase internal
43
NMA111-CYT
Protein
Nuclear mediator of apoptosis in cytososl
44
NMA111-NUC
Protein
Nuclear mediator of apoptosis in nucleus
45
NUC1-MT
Protein
NUClease 1 in mitochondria
46
NUC1-NUC
Protein
NUClease 1 in nucleus
47
PKA
Protein
Protein kinase A
48
POR1-2
Protein
PORin
49
PROTOSOM
Complex
PROTOSOM
50
PTP2
Protein
Protein tyrosine phosphatase
(Continued)
www.frontiersin.org
December 2012 | Volume 3 | Article 446 | 3
Kazemzadeh et al.
Boolean model of yeast apoptosis
Table 1 | Continued
ID
Species
Type
Name description
51
PTP3
Protein
Protein tyrosine phosphatase
52
RAS2
Protein
Homologous to RAS proto-oncogene
53
RedActinDyn
Change
Reduced actin dynamic
54
RLM1
Protein
Resistance to lethality of MKK1P386 overexpression
55
ROS-CYT
Molecule
Reactive oxygen species in cytososl
56
ROS-MT
Molecule
Reactive oxygen species in mitochondria
57
RPD3
Protein
Reduced potassium dependency
58
SALT
Input
–
59
SDP1
Protein
Stress-inducible dual specificity phosphatase
60
SLT2
Protein
SYNtaxin (SYN8)
61
SNO1
Protein
SNZ proximal open reading frame
62
SOD1
Protein
Superoxide dismutase
63
SOD2
Protein
Superoxide dismutase
64
SRO7
Protein
Suppressor of rho3
65
STE20-CYT
Protein
Sterile in cytosol
66
STE20-NUC
Protein
Sterile in nucleus
67
STM1-CYT
Protein
Translation initiation factor (TIF3) in cytososl
68
STM1-NUC
Protein
Translation initiation factor (TIF3) in nucleus
69
STRESS
Input
Event
70
SVF1
Protein
SurVival factor
71
TAT-D
Protein
3′ →5′ exonuclease and endonuclease
72
TOR1
Protein
Target of rapamycin
73
YCA1
Protein
MetaCAspase
Table includes name, type, and description of each species involved in yeast apoptosis.
*Species refers to all types of nodes that are depicted in the network map and are color coded based on their location or activity in cell.
**Housekeeping function refers to those genes which are present and give snapshot of state of the cell before applying any kind of treatment.
Table 2 | Summary of the interactions without successor
(Sink/Output) and predecessor (Source/Input).
Species
Type of connection
Number of connections
Ros-MT
Sink/output
1
MCD1-NUC
Sink/output
1
H2B
Sink/output
5
CAMP
Sink/output
2
RedActinDyn
Source/input
3
AbnormalTelomer
Source/input
1
HK
Source/input
18
Table contains species without successor (there are no edges coming out of these
nodes) and predecessor (there are no edges going to these nodes).
PREDICTIONS WITH THE CONTINUOUS MODEL
Based on qualitative knowledge we have constructed a discrete
Boolean model of yeast apoptosis. While it can capture its essen-
tial behavior,the question remained how this model can be used to
predict the qualitative behavior of the system. In order to address
this question we expanded the model by transforming the discrete
Boolean model into a continuous model using Odefy (Wittmann
et al., 2009; Krumsiek et al., 2010; see Materials and Methods).
In order to test the continuous model and its predictive capac-
ity, we performed three independent case-studies: (i) induction
of apoptosis by activation of Hog1p by heat stress, (ii) inhibition
by Bir1p of acetic acid-induced apoptosis, and (iii) induction of
apoptosis with H2O2 by activation of Stm1p. We show here that
the behavior that emerges from specific interactions in the model
is in agreement with published experimental data.
ACTIVATION OF HEAT STRESS-INDUCED APOPTOSIS WITH Hog1p
We simulated the activation of the mitogen-activated protein
kinase that is a key component of the HOG pathway (Albertyn
and Hohmann, 1994; Van Wuytswinkel et al., 2000; Hohmann,
2002; de Nadal et al., 2004), which is an osmoregulatory sig-
nal transduction cascade (Hohmann, 2009). Upon stress, Hog1p
(encoded by HOG1/YLR113W) regulates the expression of almost
600 genes by phosphorylating several different transcription fac-
tors (Hohmann, 2002; Westfall et al., 2004). The activity and
nuclear localization of Hog1p is regulated by its phosphorylation
state,and that in turn is regulated by the kinase MAPKK Pbs2p and
the phosphatases Ptc1p, Ptc2p, Ptc3p, Ptp2p, and Ptp3p (Brewster
et al., 1993; Ferrigno et al., 1998; Warmka et al., 2001; Young et al.,
2002). Besides the induction by osmotic stress, the HOG path-
way can be induced by heat stress, via a Sho1p-dependent sensory
mechanism (Winkler et al., 2002), thus we used heat stress as an
input to activate the HOG pathway and simulate the cell death
response. Heat stress activates Slt2 which then activates Rlm1.
Consequently Rlm1 triggers expression of Slt2 and PTP2 form-
ing a feedback loop. On the other hand heat shock inhibits activity
Frontiers in Physiology | Systems Biology
December 2012 | Volume 3 | Article 446 | 4
Kazemzadeh et al.
Boolean model of yeast apoptosis
FIGURE 1 | Schematic representation ofYeast Apoptosis network. Blue boxes depict input nodes, yellow nodes are placed in mitochondria, orange nodes
reside in cytosol and grey nodes belong to nucleus. Green arrows show activation effect and red arrows show inhibition effect. Blue circles depict “AND” gate.
of PKA leading to release the inhibition effect of PKA on MSN2-
4. MSN2-4 activates Sdp1 which then along with PTP2 inhibit
Slt2. We started by setting all other stimuli to zero and then by
transforming the Boolean apoptosis model into Hill Cube contin-
uous model we performed the simulations. Steady states predicted
from continuous Hill Cube (Figure 2) and synchronous Boolean
(Figure 3) model are in perfect agreement with each other for all
nodes apart from the nodes corresponding to the feedback loops
in response to heat and osmotic stress. Feedback loop in heat acti-
vated pathway includes activation of Rlm1 by Slt2 and expression
of Slt2 by Rlm1. Osmotic shock induced Hog1 activates Rlm1
to regulate Slt2 which was inhibited by osmotic shock and PTP2.
Unknown Hog1 dependent transcription factor triggers transcrip-
tion of PTP3 and PTP2 resulting in dephosphorylation of Hog1
phosphotyrosine which inhibits Hog1 activities. All nodes in our
model were connected to activation of apoptosis via the heat shock
pathway and were indeed activated in our simulation and are col-
ored in red; other pathways are colored in blue indicating that they
are not activated during the simulation (Figure 2). Model predicts
that upon heat induction, concentration of Hog1p changes trough
time never reaching its maximum level of concentration and as an
intensity of stimulus decreases level of Hog1p also decreases. Our
simulations also proposed that the maximal level of activation of
Hog1p during heat stress is 70% of the total activation and as the
heat stimulus continues over time its activity decreases and reaches
a plateau at 40% of total activation (Figure 4).
This model prediction is in agreement with an experimental
measurement performed by Winkler et al. (2002) which shows the
induced activity of Hog1p upon heat induction (Figure 4). The
difference between the maximal activity values and the lag phase of
the activation is due to the fact that we did not use any quantitative
data as input to the model.
INHIBITION OF ACETIC ACID-INDUCED APOPTOSIS WITH Bir1p
Even though there are several genes and pathways that are involved
in yeast pro-apoptotic response, there are only few anti-apoptotic
genes. For many years Bir1p (encoded by BIR1/YJR089W) was
thought to be the only apoptotic inhibitor in yeast, but recently
a TSC22-protein family was found and two of the proteins
with TSC22-motif have been shown to have anti-apoptotic roles
Sno1p and Fvy10p (Khoury et al., 2008). Bir1 belongs to the
IAP family and phylogenetic analysis (Uren et al., 1998) showed
similarity to Schizosaccharomyces pombe BIR1, human survivin,
and Caenorhabditis elegance BIR-1 and BIR-2 proteins. Yeast’s
www.frontiersin.org
December 2012 | Volume 3 | Article 446 | 5
Kazemzadeh et al.
Boolean model of yeast apoptosis
Table 3 | List of logical interactions in yeast Boolean model.
ID
Interaction
Function
Time scale
Reference
1
HK =AIF1-MT
Housekeeping
0
2
HK = DRE2/TAH18
Housekeeping
0
3
HK = EMC4
Housekeeping
0
4
HK = SVF1
Housekeeping
0
5
HK = FVY10
Housekeeping
0
6
HK = SOD2
Housekeeping
0
7
HK = SNO1
Housekeeping
0
8
HK = NDI1
Housekeeping
0
9
HK = POR1-2
Housekeeping
0
10
HK = MMI1
Housekeeping
0
11
HK = MCD1-MT
Housekeeping
0
12
HK = SRO7
Housekeeping
0
13
HK = CDC48
Housekeeping
0
14
HK = FIS1
Housekeeping
0
15
HK = MDV1
Housekeeping
0
16
HK = STM1-CYT
Housekeeping
0
17
=AceticAcid
Input
2
18
=Adozelesin
Input
2
19
=CU2
Input
2
20
=H2O2
Input
2
21
=Mating
Input
2
22
=MG2
Input
2
23
=Salt
Input
2
24
=Heat
Input
2
25
=Stress
Input
2
26
!SOD2 + NDI1 = ROS-MT
4
Li et al. (2006)
27
AceticAcid = CytC-MT
4
Ludovico et al. (2002)
28
CDC48 = CytC-CYT
4
Eisenberg et al. (2007)
29
CytC-CYT =YCA1
4
Eisenberg et al. (2007)
30
CytC-MT = CytC-CYT
4
Eisenberg et al. (2007)
31
MCD1-MT = CytC-MT
4
Yang et al. (2008)
32
MEC1 =YCA1
4
Weinberger et al. (2005)
33
MT-Frag = MT-ALT
4
Wissing et al. (2004)
34
!FYV10 =Apoptosis
4
Khoury et al. (2008)
35
2 CDC48 = ROS-CYT
4
–
36
CU2 + CPR3 =Apoptosis
4
Liang and Zhou (2007)
37
DNA-Frag =Apoptosis
4
Madeo et al. (2009)
38
ESP1 = ROS-CYT
4
Yang et al. (2008)
39
MT-Frag =YCA1
4
Eisenberg et al. (2007)
40
NMA111-CYT = NMA111-NUC
4
Walter et al. (2006)
41
NUC1-MT = KAP123
4
Buttner et al. (2007)
42
RAS2 =AdenylateCyclase
4
Wood et al. (1994)
43
RAS2 = ROS-CYT
4
Kataoka et al. (1984)
44
RedActinDyn = ROS-CYT
4
Eisenberg et al. (2007)
45
RedActinDyn =YCA1
4
Madeo et al. (2009)
46
ROS-CYT =Apoptosis
4
Eisenberg et al. (2007)
47
ROS-CYT =YCA1
4
Madeo et al. (2002)
48
Salt = ROS-CYT
4
Wadskog et al. (2004)
49
SOD1 = ROS-CYT
4
Eisenberg et al. (2007)
50
STE20-NUC = H2B
4
Madeo et al. (2009)
(Continued)
Frontiers in Physiology | Systems Biology
December 2012 | Volume 3 | Article 446 | 6
Kazemzadeh et al.
Boolean model of yeast apoptosis
Table 3 | Continued
ID
Interaction
Function
Time scale
Reference
51
Stress = RPD3
4
Ahn et al. (2006)
52
Tat-D = DNA-Frag
4
Qiu et al. (2005)
53
!SNO1 =Apoptosis
4
Khoury et al. (2008)
54
AbnormalTelomer = MEC1
4
Weinberger et al. (2005)
55
AdenylateCyclase = CAMP
4
Schmelzle et al. (2004)
56
Adozelesin = CDC6
4
Blanchard et al. (2002)
57
AIF1-MT =AIF1-NUC
4
Wissing et al. (2004)
58
AIF1-NUC = H2B
4
Wissing et al. (2004)
59
Apoptosis =
Output
4
–
60
CDC6 = Protosom
4
Blanchard et al. (2002)
61
ESP1 = MCD1-NUC
4
Yang et al. (2008)
62
H2O2 = NUC1-MT
4
Buttner et al. (2007)
63
H2O2 = ESP1
4
Yang et al. (2008)
64
H2O2 = HOS3
4
Carmona-Gutierrez et al. (2010a)
65
Heat = NMA111-CYT
4
Walter et al. (2006)
66
HOS3 = H2B
4
Carmona-Gutierrez et al. (2010a)
67
KAP123 = NUC1-NUC
4
Buttner et al. (2007)
68
MAPK = STE20-CYT
4
Carmona-Gutierrez et al. (2010a)
69
MatingPheromone = MAPK
4
Carmona-Gutierrez et al. (2010a)
70
Mg2+ =Tat-D
4
Qiu et al. (2005)
71
MMI1 = MT-ALT
4
Eisenberg et al. (2007)
72
MT-ALT = MT-FRAG
4
–
73
NUC1-NUC = H2B
4
Buttner et al. (2007)
74
PKA = MT-ALT
4
Carmona-Gutierrez et al. (2010a)
75
RAS2 = MT-ALT
4
Eisenberg et al. (2007)
76
RAS2 = PKA
4
Carmona-Gutierrez et al. (2010a)
77
RedActinDyn = RAS2
4
Eisenberg et al. (2007)
78
RPD3 = H2B
4
Ahn et al. (2006)
79
STE20-CYT = STE20-NUC
4
Carmona-Gutierrez et al. (2010a)
80
Stress =AdenylateCyclase
4
Schmelzle et al. (2004)
81
TOR1 = CAMP
4
Schmelzle et al. (2004)
82
TOR1 = RAS2
4
Schmelzle et al. (2004)
83
Heat = SOD1
4
84
2 NDI1 = ROS-CYT
4
–
85
Stress =TOR1
4
Schmelzle et al. (2004)
86
!PTP2 = SLT2
5
Hahn and Thiele (2002)
87
!PTP2 = HOG1
5
Hahn and Thiele (2002)
88
HOG1 = HOG1-Dep
5
Hahn and Thiele (2002)
89
!PTP3 = HOG1
5
Hahn and Thiele (2002)
90
!SDP1 = SLT2
5
Hahn and Thiele (2002)
91
!SLT2 = DesCyclinCCDK8
5
Krasley et al. (2006)
92
!Stress = SLT2
5
Hahn and Thiele (2002)
93
DesCyclinCCCDK8 = ROS-CYT
5
Krasley et al. (2006)
94
Heat = PKA
5
Hahn and Thiele (2002)
95
Heat = SLT2
5
Hahn and Thiele (2002)
96
Hog1 = RLM1
5
Hahn and Thiele (2002)
97
HOG1-Dep = PTP3
5
Hahn and Thiele (2002)
98
MSN2-4 = SDP1
5
Hahn and Thiele (2002)
99
PKA = MSN2-4
5
Hahn and Thiele (2002)
100
RLM1 = PTP2
5
Hahn and Thiele (2002)
101
RLM1 = SLT2
5
Hahn and Thiele (2002)
(Continued)
www.frontiersin.org
December 2012 | Volume 3 | Article 446 | 7
Kazemzadeh et al.
Boolean model of yeast apoptosis
Table 3 | Continued
ID
Interaction
Function
Time scale
Reference
102
SLT2 = RLM1
5
Hahn and Thiele (2002)
103
Stress = HOG1
5
Hahn and Thiele (2002)
104
!SRO7 + Salt =YCA1
6
Wadskog et al. (2004)
105
!STM1-NUC = DNA-Frag
6
Ligr et al. (2001)
106
AceticAcid + !SVF1 = ROS-CYT
6
Vander Heiden et al. (2002)
107
H2O2 + !EMC4 = ROS-CYT
6
Ring et al. (2008)
108
H2O2 + !SVF1 = ROS-CYT
6
Vander Heiden et al. (2002)
109
!FIS1 + MDV1 = MT-Frag
6
Eisenberg et al. (2007)
110
!POR1-2 +AceticAcid =Apoptosis
6
Pereira et al. (2007)
111
!POR1-2 + H2O2 =Apoptosis
6
Pereira et al. (2007)
112
H2O2 + !DRE2/TAH18 = MT-Frag
6
Vernis et al. (2009)
113
YCA1 + !BIR1 =Apoptosis
6
Walter et al. (2006)
114
!NMA111-NUC = BIR1
6
Walter et al. (2006)
115
STM1-CYT + !Protosom = STM1-NUC
6
Ligr et al. (2001)
Table includes involved species in each interaction, logical rule for each interaction and time scale in which interaction takes place. Note: the interactions of the model
are given in the notation of the Cell Net Analyser: a logical NOT is represented by “!”; a logical AND is represented by “+” and interaction on right hand side of “=”
gives the value of node on left hand side or equation.
Table 4 | Predicted states of relevant species at steady state.
Species
t = 0
t = 4
t = 5
t = 6
Acetic acid
1
1
1
1
Adozelesin
1
1
1
1
AIF1-MT
1
1
1
1
AIF1-NUC
0
1
1
1
Apoptosis
0
0
0
1
BIR1
0
0
0
0
DNA-FRAG
0
1
1
1
H2O2
1
1
1
1
Heat
1
1
1
1
NMA111-NUC
0
1
1
1
ROS-CYT
0
1
1
1
STE20-CYT
0
1
1
1
STM1-CYT
1
1
1
0
STM1-NUC
0
0
0
1
YCA1
0
1
1
1
Table includes time scale scenario upon induction of all stimuli.T stands for differ-
ent time steps. As an example if Acetic Acid is inducted to the cell at time point 0
it is expected to cause DNA Fragmentation at next time step and eventually ends
to apoptosis in last step.
Bir1p has been previously intensively studied in chromosome seg-
regation (as a component of the Aurora kinase complex (chromo-
some passenger complex, CPC; Ruchaud et al., 2007) but recently
studies of its role as a negative regulator of apoptosis have gained
momentum. It has been shown that Bir1p is a target for degrada-
tion by Nma111p (Owsianowski et al., 2008), an apoptotic serine
protease and yeast cells lacking Bir1 are more sensitive to apopto-
sis, while overexpression of Bir1 reduces apoptosis (Walter et al.,
2006).
When building the model we assumed that addition of
acetic acid induces cytochrome c release from mitochondria
and its translocation to the cytosol. Another assumption is that
the execution of apoptosis is Yca1p-dependent (encoded by
YCA1/MCA1/YOR197W) in this context and that the activation
of the caspase is downstream from the cytochrome c transloca-
tion (Ludovico et al., 2001, 2002). Thus, taking these assumptions
into account acetic acid was used as an inducer in the simula-
tions with direct induction of apoptosis. The model confirmed
experimental evidence that Bir1 is indeed apoptosis inhibitor in
yeast (Walter et al., 2006) and as acetic acid is applied as a pulse
stimulus (that is then decreasing over time) and cytochrome c
present in the cytosol increases, the decrease of Bir1p due to
Nma111p degradation promotes apoptosis that then reaches the
maximum (Figure 5A). We then performed an artificial and bio-
logically irrelevant simulation: after apoptosis has occurred and
no more inducer was present (acetic acid is not added again and
the previously added amount has been “used”) the eventual the-
oretical accumulation of Bir1p did inhibit apoptosis and revert it
to zero. Obviously this scenario is biologically implausible, since
once the cell has undergone apoptosis it cannot be revived, but
has a purpose to show mathematical correctness of the developed
model.
Additionally, we have converted discreet apoptosis model to
continuous model with constant presence of Bir1 in order to
test if this type of model would predict the same outcome as
the experimental approach in which Bir1p is overexpressed and
provides protection from induction of apoptosis. The continu-
ous model was created taking into account translocation effect
of each element from one compartment to another. This effect
has been applied to translocation of Nuc1 from mitochondria to
nucleus, MCD1 from nucleus to mitochondria, Stm1 from cytosol
to nucleus Aif1 (from mitochondria to nucleus, NMA111 from
cytosol to nucleus, Ste20 from cytosol to nucleus, and cytochrome
Frontiers in Physiology | Systems Biology
December 2012 | Volume 3 | Article 446 | 8
Kazemzadeh et al.
Boolean model of yeast apoptosis
HillCube
Heat
H202
Mg2
Adozelesin
Acetic Acid
CU2
Salt
Mating
Stress
AIF1-MT
CYTC-MT
MMI1
CPR3
DRE2-Tah18
FIS1
MDV1
SOD2
ROS-MT
NUC1-MT
SVF1
CDC6
EMC4
POR1-2
NMA111-CYT
NMA111-NUC
PROTOSOM
STM1-CYT
STM1-NUC
DNA-FRAG
Tat-D
SNO1
CYTC-CYT
KAP123
NUC1-NUC
CDC48
ROS-CYT
YCA1
SOD1
NDI1
MEC1
MAPK
PKA
STE20-CYT
MT-FRAG
MT-ALT
TOR1
Adelynatecyclase
RAS2
RedActionDyn
RPD3
STE20-NUC
ESP1
MCD1-NUC
AIF1-NUC
HOS3
FVY10
H2B
BIR1
Apoptosis
AbnormalTelomer
MCD1-MT
SRO7
CAMP
HK
MSN2-4
SDP1
SLT2
RLM1
PTP2
PTP3
HOG1_Dep
HOG1
DesCyclinCCDK8
Species
Time [arbitrary units]
0
FIGURE 2 | Continuous Hill Cube transformation. All species in apoptosis network are mapped on vertical axis. Dark blue color indicates those nodes that are
not activated (value = 0) while dark red refers to nodes which are completely activated (value = 1). Each color in between indicates the level of activation
between 0 and 1.
c from mitochondria to cytosol. Initially, in this model, translo-
cation of each species is modeled as one node in order to rep-
resent the re-localization which introduced self-regulatory loops
to the system which is impossible since to our knowledge yeast
apoptosis regulatory network does not have such loops. One
solution implies intuitive approach in defining single variable
for single species to mimic the effect of transferring from one
compartment to another. Technically this was solved by intro-
ducing two variables for a single species representing both com-
partments they can belong to. This approach, also, confirmed
experimental finding (Walter et al., 2006) that constant pres-
ence of Bir1, inhibits apoptosis, validating Bir1 anti-apoptotic role
(Figure 5B).
ACTIVATION OF H2O2-INDUCED APOPTOSIS WITH Stm1p
It has been shown that degradation of short-lived pro-apoptotic
proteins via proteasomal – ubiquitine pathway plays important
role in mammalian apoptosis (Drexler, 1997). One of the yeast
proteasomal substrates – Stm1 (YLR150W) was identified in the
study by Ligr et al. (2001) as an activator of the cell death process.
Conservedorthologsof Stm1aredetectedinseveralspecies(highly
conserved ortholog in Schizosaccharomyces pombe and a putative
ortholog in Drosophila melanogaster (Nelson et al., 2000) suggest-
ingthatregulationof apoptosisviathisproteincanbeevolutionary
conserved process.
We have in silico tested two scenarios confirming the role of
Stm1 in apoptosis (Ligr et al., 2001). Accumulation of Stm1 will
induce apoptotic behavior followed by DNA fragmentation which
is a known marker of cell death, while the Stm1 knock out will
promote survival and consequently deterring DNA fragmentation
(Figures 6A,B).
IN SILICO HUMANIZED YEAST APOPTOSIS
S. cerevisiae is a model organism that has conserved genes, pro-
teins and pathways that are similar to the ones in the human cells.
This allows for using yeast as a host in which human genes can be
expressed, so called “humanized yeast” (Winderickx et al., 2008)
and subsequently the physiological roles and molecular mecha-
nisms can be studied. In order to test if our system could be used
in a similar way (in silico humanized yeast) and provide reliable
simulations and predictions, we inserted three human genes with
complete downstream pathways into our initial yeast apoptosis
www.frontiersin.org
December 2012 | Volume 3 | Article 446 | 9
Kazemzadeh et al.
Boolean model of yeast apoptosis
Heat
H202
Mg2
Adozelesin
Acetic Acid
CU2
Salt
Mating
Stress
AIF1-MT
CYTC-MT
MMI1
CPR3
DRE2-Tah18
FIS1
MDV1
SOD2
ROS-MT
NUC1-MT
SVF1
CDC6
EMC4
POR1-2
NMA111-CYT
NMA111-NUC
PROTOSOM
STM1-CYT
STM1-NUC
DNA-FRAG
Tat-D
SNO1
CYTC-CYT
KAP123
NUC1-NUC
CDC48
ROS-CYT
YCA1
SOD1
NDI1
MEC1
MAPK
PKA
STE20-CYT
MT-FRAG
MT-ALT
TOR1
Adelynatecyclase
RAS2
RedActionDyn
RPD3
STE20-NUC
ESP1
MCD1-NUC
AIF1-NUC
HOS3
FVY10
H2B
BIR1
Apoptosis
AbnormalTelomer
MCD1-MT
SRO7
CAMP
HK
MSN2-4
SDP1
SLT2
RLM1
PTP2
PTP3
HOG1_Dep
HOG1
DesCyclinCCDK8
Time [arbitrary units]
1
Boolean (synchronous)
Species
FIGURE 3 | Synchronous Boolean model. All species in apoptosis network are mapped on vertical axis. Dark blue indicates those nodes that are not activated
(value = 0) while dark red refers to nodes which are totally activated (value = 1).
Boolean model. Yeast apoptotic network was “humanized” by
in silico insertion of genes belonging to Bcl-2 protein family
and Valosin-contain protein (VCP) – distinctive representatives
of human apoptosis (Figure 7).
INSERTION OF Bcl-2 FAMILY PROTEINS: ANTI-APOPTOTIC Bcl-xL AND
PRO-APOTOTIC BAX
This protein family consists of apoptotic agonist and anti-agonist
proteins. Among members of Bcl-2 family Bax as an apoptotic
inducer and Bcl-xL as an anti-apoptotic factor were included in
our extended model. Bax is located in cytosol in mammalian cells
and has vital role in mitochondria morphogenesis. Heterologously
expressed Bax causes growth arrest and rapid cell death in S. cere-
visiae (Sato et al., 1994; Greenhalf et al., 1996). Also expression
of Bax has been linked to the release of cytochrome c from mito-
chondria (Manon et al., 1997). Yeast cells with Bax expression
accumulate ROS and show other apoptotic hallmarks such as DNA
fragmentation (Madeo et al., 1999).
Since members of this protein family were previously success-
fully expressed in yeast, we first validate the “humanized” yeast
model by inserting Bcl-xL, Bax, Bad, and Bcl-2 genes (Sato et al.,
1994; Greenhalf et al., 1996; Madeo et al., 1999). As an input signal
human Akt signaling was used (Akt Signaling), as an extrinsic
regulatory switch (since this signaling cascade is not present in
yeast). The Akt Serine/Threonine-kinase promotes cell survival by
phosphorylating the pro-apoptotic protein BAD (member of the
Bcl-2 family), which is the cause of dissociation of BAD from the
Bcl-2/Bcl-X complex, and promotion of cell survival (Datta et al.,
1997). Besides cell survival,Akt signaling is related to the cell cycle,
metabolism,and angiogenesis and therefore a target for anticancer
drug development (Falasca, 2010; Hers et al., 2011).
Upon activation of Akt Signaling at the first time step (t = 0,
node value Akt Signaling = 1), Bcl-2 and Bcl-xL are activated in
the following step (t = 4, node value Bcl-xL = 1 and Bcl-2 = 1)
and remain active throughout the simulation and as the inhibitors
of apoptosis promote the survival (node value Apoptosis = 0;
Table 5). Simulation results suggest the anti-apoptotic role of two
Bcl-2 family members: Bcl-2 and Bcl-xL which is consistent with
experimental findings (Kharbanda et al., 1997).
DUAL FUNCTIONALITY OF VCP IN SURVIVAL AND APOPTOSIS
The evolutionary conserved Valosin-containing protein (VCP) is
a mammalian ortholog of yeast Cdc48, which is the first apoptotic
mediator found in S. cereivisae (Braun and Zischka, 2008). VCP is
Frontiers in Physiology | Systems Biology
December 2012 | Volume 3 | Article 446 | 10
Kazemzadeh et al.
Boolean model of yeast apoptosis
the member AAA-ATPase family which is ubiquitously expressed
(Braun and Zischka,2008). It consists of four domains:N-terminal
domain, two ATPase domains D1 and D2 and C-terminal domain
(Wang et al., 2004). The major ATPase activity of VCP is carried
FIGURE 4 | Hog1 study. Comparison of experimental and simulation study.
Black curve shows activity of wild type Hog1 upon induction of heat
(Winkler et al., 2002) and red curve illustrates Hog1 activation level in
continues simulation.
on by D2 domain (Wang et al., 2004; Song et al., 2003). VCP is
involved in different cellular process such as protein degradation,
membrane fusion and chaperone activity (Braun and Zischka,
2008). Role of VCP/Cdc48 in fluctuating number of death cell
in various types of disease naming cancer and protein deposition
diseases is not well understood. Increase in expression of VCP
is correlated to the development of cancer and metastasis there-
fore detecting the level of VCP expression is proposed as cancer
progression marker. Also VCP is known as detector of aggregated
proteins causing neurodegenerative disease such as Parkinson and
Alzheimer (Hirabayashi et al., 2001; Mizuno et al., 2003; Ishigaki
et al., 2004). It has been observed that yeast cell carrying mutation
of Cdc48 is showing morphological markers of apoptosis (Madeo
et al., 1997) and that depletion of this protein has apoptotic effect
in other organisms as well (Imamuraab et al., 2003).
It had been shown that VCP can both promote or inhibit apop-
tosis (Braun and Zischka,2008). Deletion of VCP triggers ER stress
which is followed by unfolded protein response and cell death via
ER-associated degradation (ERAD) pathway – a highly conserved
pathway between mammalian and yeast cells (Braun and Zischka,
2008). Wild type VCP has pro-apoptotic role in the cells undergo-
ing apoptosis upon ER stress which in turn triggers ER-associated
apoptotic pathway. Finally VCP can trigger survival pathway in
response to NFkB which is a pro-survival molecule in cells with
over expressed level of VCP/Cdc48(Braun and Zischka, 2008).
The dual functionality of VCP in survival and apoptosis mech-
anism makes it an attractive candidate to be inserted in initial
yeast apoptosis Boolean network. Since VCP includes expression
of a handful of heterologous proteins, its in vivo insertion to
yeast plasmid would be difficult to perform and thus represents a
FIGURE 5 | Bir1 study. (A) Acetic acid (green) is applied as a pulse stimulus (that is then decreasing over time) and cytochrome c (yellow) present in the cytosol
increases, the decrease of Bir1p (red) promotes apoptosis (B) Constant presence of Bir1p (red) inhibits apoptosis (black), validating Bir1 anti-apoptotic role.
www.frontiersin.org
December 2012 | Volume 3 | Article 446 | 11
Kazemzadeh et al.
Boolean model of yeast apoptosis
FIGURE 6 | Stm1 study. (A) Presence of Stm1p (blue) promotes apoptosis (yellow; B) Knock out of Stm1p (blue) prevents apoptosis (yellow) and DNA
fragmentation (red) and consequently promotes survival.
FIGURE 7 | In silico “humanized yeast apoptosis network”. Human apoptotic pathways BCL-2 protein family and VCP dependent genes inserted into the
yeast apoptosis network (due to simplicity we show only human pathways).
Frontiers in Physiology | Systems Biology
December 2012 | Volume 3 | Article 446 | 12
Kazemzadeh et al.
Boolean model of yeast apoptosis
Table 5 | Summary of simulations upon insertion of Bcl-2 pathway.
Species
t = 0
t = 4
t = 5
t = 6
UV
0
0
0
0
Akt signaling
1
1
1
1
BCL-XL
0
1
1
1
P53
0
0
0
0
BAD
0
0
0
0
BAX
0
0
0
0
BCL-2
0
1
1
1
Apoptosis
0
0
0
0
Table includes simulation results of heterologous expression of BCL-2 in yeast
apoptosis model.T stands for different time steps. Upon activation of Akt Signal-
ing pathway at first time step BCL-2 gets activated in following step and as an
inhibitor of apoptosis prevents apoptosis till end point.
good candidate for further investigation and exploration of model
capabilities.
In our model, yeast Cdc48 was replaced with human VCP gene
and its downstream pathway effector caspases (caspases 3, 6 and
7), initiator caspases (caspase9 and 12), IAP family, IkB-alpha
inhibitory protein which when degraded by proteasome cause the
release, and nuclear translocation of active NFkB (represented in
the model as NFkB_Cyt and NFkB_Nuc) and gp130/Stat3 pathway
(represented as a single node).
Upon activation of Akt Signaling pro-survival role of VCP is
observed (node Survival = 1 at t = 6, Table 3). This is achieved
when IkBα is dissociated from NFkB (node NFkB/IkBα = 1 at
t = 4), which in turn gets activated (node NFkB = 1, t = 5)
and is translocated to the nucleus (nodes NFkB_Cyt = 1 and
NFkB_Nuc = 1, t = 6) inducing survival of the cell (node Sur-
vival = 1, t = 6). Simultaneously, activation of NFkB, promotes
activity of IAP family of proteins which inhibits the activity of both
effector and initiator caspases (nodes c-3-6-7 = 0 and c-9-12 = 0,
t = [0,4,5,6],thusdisablingapoptosis-dependentcaspasepathway.
As an independent, but parallel process, activation of VCP via
Akt Signaling activates gp130-Stat3 (node gp130-Stat3 = 1, t = 4)
pathway and consequently leads to survival (Table 6).
This result suggests that yeast carrying mammalian VCP gene
exhibits the same behavior as it is known from the human
apoptotic model (Vandermoere et al., 2006).
DISCUSSION
In this work we have constructed a Boolean model for the bio-
chemical network that controls apoptosis pathway in budding
yeast Saccharomyces cerevisiae. Firstly, we presented the Boolean
model describing only yeast pro and anti-apoptotic genes and val-
idated the model by further analyzing the role of Stm1p, Bir1p,
and Hog1p. Even though construction of the yeast apoptosis net-
work involved certain simplifications (nodes have only two states
and rules are describing network dynamics) we were able to model
more complex network than using dynamic modeling approach
that requires knowledge of kinetic parameters for all molecular
processes. This approach was able to suggest general design prin-
cipals of yeast apoptosis. One of the advantages of constructing
mathematical models in biology is that we are able to simulate
Table 6 | Summary of simulations upon insertion of VCP pathway.
Species
t = 0
t = 4
t = 5
t = 6
Akt signaling
1
1
1
1
GP130-STAT3
0
1
1
1
NFkB-CYT
0
1
1
1
NFkB-NUC
0
1
1
1
NFkB/IkBα
0
1
1
1
VCP
1
1
1
1
C-3-6-7
0
0
0
0
C-9-12
0
0
0
0
IkBα
0
1
1
1
IAP
0
1
1
1
Survival
0
0
0
1
Apoptosis
0
0
0
0
Table includes simulation results of humanized yeast apoptosis model by inser-
tion of VCP. T stands for different time steps. VCP in presence of Akt Signaling is
expected to prevent apoptosis and promote survival at last time point.
scenarios that are not feasible in real experiments. In the case
of Bir1 p we were able to in silico revive a cell already reaching
apoptosis. These examples are important to understand general
mechanisms of the constructed network. A study of proteasomal
substrate Stm1p suggested that Stm1p seems to be an effector of
H2O2 and the presence of Stm1p with an inducer leads to apopto-
sis in our model,and in Stm1p knock out survival is promoted and
DNA fragmentation is avoided, irrespective of the presence of the
inducer. Despite the limitation of BN in terms of giving quanti-
tative predictions of the system dynamics they allow investigation
of the large networks and their systematic exploration resulting
in better understanding of cellular processes. Example of Hog1p
study showed that our model was capable of reproducing the pat-
tern of Hog1p activation,but was not able to quantitatively predict
the maximal Hog1p activity.
In the second stage, we extended the initial model by inserting
two pathways of human apoptosis (VCP and Bcl-2 family), hereby
creating a“humanized”in silico yeast. Humanized yeast strains are
used in experimental molecular biology where the human proteins
(causing, or associated with diseases) are expressed and studied
in vivo (in yeast in this case).
Conservation of many apoptotic mediators and mechanisms
among the Eukarya provides the possibility of inserting genes
from other organisms into yeast (in vivo) or in the yeast apop-
totic network (in silico). We validated constructed humanized
yeast model by insertion of Bcl-2 family which have been pre-
viously successfully expressed in yeast. The model was able to
reproduce well-known pro and anti-apoptotic phenotypes con-
firming that yeast expressing Bax accumulated ROS and showed
other apoptotic markers like DNA fragmentation. To test whether
or not specific pathways playing role in neurodegeneration and
cancer can be elucidated by our Boolean model and can we con-
sequently take advantage of a simple system like yeast to explore
hypothesis generated for higher organisms, we in silico expressed
evolutionary conserved VCP and its downstream components in
existing yeast apoptosis network. VCP is important player in can-
cer cell survival and can be used as a target for cancer therapy. It
www.frontiersin.org
December 2012 | Volume 3 | Article 446 | 13
Kazemzadeh et al.
Boolean model of yeast apoptosis
also serves as detector of aggregated proteins that are known to
be cause of neurodegenerative diseases such as Parkinson’s and
Alzheimer’s. Our model showed that cell survival is mediated
by VCP via the degradation of IkBα that leads to translocation
of NFkB to nucleus. This result is in agreement with experi-
mental findings that VCP is an essential target in the Akt sig-
naling pathway suggesting that presented model in combination
with experimental approach would represents a promising plat-
form to study complex cellular processes involved in cancer and
neurodegeneration.
Exploiting the advantages of BN models that enables extrac-
tion of system level properties of large networks and extensive
state exploration and converting discrete model to continuous
model our results suggested that in contrast to human apoptotic
network yeast apoptosis is linear process whose regulation does
not involve any complex feedback loops. Analysis of the second
model showed that with certain adjustments Boolean model of
yeast apoptosis can be adapted for studies of apoptosis in higher
organisms.
Our results show that even without kinetic and qualitative data,
it is possible to build models that can simulate relevant states of
yeast physiology and regulations and can contribute to further
understanding of biology. Since yeast is a preferred model organ-
ism for many studies of fundamental processes in a Eukaryal cell,
we argue that in silico studies of yeast will be an important contrib-
utor to the understanding of complex cellular regulations, such as
cell death pathways and that the applications will extend toward
study of regulations or causes of human diseases such as cancer
and neurodegeneration.
MATERIALS AND METHODS
APOPTOSIS NETWORK SETUP
Based on the extensive literature study and databases search (Sac-
charomyces Genome Database-SGD; Cherry et al.,2011) the apop-
tosis network consisting of 73 nodes and 115 edges is constructed.
These nodes are selected based on their interactions and their sub-
strates involved in apoptosis. Cell Designer (Funahashi et al.,2003)
was used to visualize genes and their connection. Cell Designer
allowed us to represent network using comprehensive graphical
notation. Moreover, Cell Designer is able to connect to online data
bases such as KEGG (Kanehisa and Goto, 2000), BioModels (Li
et al., 2010) and PubMed which expands its connectivity, visual-
izationandmodelbuilding.Themostprominentadvantageof Cell
Designer is its ability to support SBML (Systems Biology Markup
Language; Hucka et al., 2003) format. Creating SBML file avoids
creating logic rule file which is more error prone when rules have
to be defined by user in simple text and it is not in form of easily
drawing boxes.
Heat
H202
Mg2
Adozelesin
Acetic Acid
CU2
Salt
Mating
Stress
AIF1-MT
CYTC-MT
MMI1
CPR3
DRE2-Tah18
FIS1
MDV1
SOD2
ROS-MT
NUC1-MT
SVF1
CDC6
EMC4
POR1-2
NMA111-CYT
NMA111-NUC
PROTOSOM
STM1-CYT
STM1-NUC
DNA-FRAG
Tat-D
SNO1
CYTC-CYT
KAP123
NUC1-NUC
CDC48
ROS-CYT
YCA1
SOD1
NDI1
MEC1
MAPK
PKA
STE20-CYT
MT-FRAG
MT-ALT
TOR1
Adelynatecyclase
RAS2
RedActionDyn
RPD3
STE20-NUC
ESP1
MCD1-NUC
AIF1-NUC
HOS3
FVY10
H2B
BIR1
Apoptosis
AbnormalTelomer
MCD1-MT
SRO7
CAMP
HK
MSN2-4
SDP1
SLT2
RLM1
PTP2
PTP3
HOG1_Dep
HOG1
DesCyclinCCDK8
Heat
H202
Mg2
Adozelesin
Acetic Acid
CU2
Salt
Mating
Stress
AIF1-MT
CYTC-MT
MMI1
CPR3
DRE2-Tah18
FIS1
MDV1
SOD2
ROS-MT
NUC1-MT
SVF1
CDC6
EMC4
POR1-2
NMA111-CYT
NMA111-NUC
PROTOSOM
STM1-CYT
STM1-NUC
DNA-FRAG
Tat-D
SNO1
CYTC-CYT
KAP123
NUC1-NUC
CDC48
ROS-CYT
YCA1
SOD1
NDI1
MEC1
MAPK
PKA
STE20-CYT
MT-FRAG
MT-ALT
TOR1
Adelynatecyclase
RAS2
RedActionDyn
RPD3
STE20-NUC
ESP1
MCD1-NUC
AIF1-NUC
HOS3
FVY10
H2B
BIR1
Apoptosis
AbnormalTelomer
MCD1-MT
SRO7
CAMP
HK
MSN2-4
SDP1
SLT2
RLM1
PTP2
PTP3
HOG1_Dep
HOG1
DesCyclinCCDK8
FIGURE 8 | Interaction Matrix ofYeast Apoptosis Network. Each row
corresponds to single species and each column corresponds to reactions. A
red matrix element eij indicates an inhibition influence of species ion reaction
j. In contras a green filed shows an activation influence of species i on
reaction j while a blue box indicates species i gets activated in reaction j and
as it is expected the black cells indicates that species i does not participate in
reaction j. Number of interactions where the species is involved is mentioned
at the end of each row as connectivity number of each species. Number of
reactions that activates/inhibits is mentioned in brackets (Color coding:
red – inhibition, green – activation and black – no interaction influence).
Frontiers in Physiology | Systems Biology
December 2012 | Volume 3 | Article 446 | 14
Kazemzadeh et al.
Boolean model of yeast apoptosis
Heat
H202
Mg2
Adozelesin
Acetic Acid
CU2
Salt
Mating
Stress
AIF1-MT
CYTC-MT
MMI1
CPR3
DRE2-Tah18
FIS1
MDV1
SOD2
ROS-MT
NUC1-MT
SVF1
CDC6
EMC4
POR1-2
NMA111-CYT
NMA111-NUC
PROTOSOM
STM1-CYT
STM1-NUC
DNA-FRAG
Tat-D
SNO1
CYTC-CYT
KAP123
NUC1-NUC
CDC48
ROS-CYT
YCA1
SOD1
NDI1
MEC1
MAPK
PKA
STE20-CYT
MT-FRAG
MT-ALT
TOR1
Adelynatecyclase
RAS2
RedActionDyn
RPD3
STE20-NUC
ESP1
MCD1-NUC
AIF1-NUC
HOS3
FVY10
H2B
BIR1
Apoptosis
AbnormalTelomer
MCD1-MT
SRO7
CAMP
HK
MSN2-4
SDP1
SLT2
RLM1
PTP2
PTP3
HOG1_Dep
HOG1
DesCyclinCCDK8
5 ( 4/0/1)
8 ( 7 /0/1)
2 (1/0/1)
2 (1/0/1)
4 ( 3/0/1)
2( 1/0/1)
3 ( 2/0/1)
2 ( 1/0/1)
6 ( 4/1/1)
2 (1/0/1)
3 (1/0/2)
2 (1/0/1)
2 (1/0/1)
2 (0/1/1)
2 (0/1/1)
2 (1/0/1)
2 (0/1/1)
1 (0/0/1)
2 (1/0/1)
3 (0/2/1)
2 (1/0/1)
2 (0 /1/ 1)
3 (0/2/1)
2 (1/0/1)
2 (0/1/1)
2 (0/1/1)
2 (1/0/1)
2 (0/1/1)
3 (1/0/2)
2 (1/0/1)
2 (0/1/1)
3 (1 /0/2)
2 (1/0/1)
2 (1/0/1)
3 (2/0/1)
13 (2/0/11)
7 (1/0/6)
2 (1/0/1)
3 (2/0/1)
2 (1/0/1)
2(1/0/1)
4 (2/0/2)
2 (1/0/1)
4 (1/0/3)
4 (1/0/3)
3 (2/0/1)
3 (1/0/2)
6 (4/0/2)
3 (3/0/0)
2 (1/0/1)
2 (1/0/1)
3 (2/0/1)
1 (0/0/1)
2 (1/0/1)
2 (1/0/1)
2 (0/1/1)
5 (0/0/5)
2 (0/1/1)
9 (1/0/8)
1 (1/0/0)
2 (1/0/1)
2 (0/1/1)
2 (0/0/2)
16 (16/0/0)
2 (1/0/1)
2 (0/1/1)
7 (1/1/5)
4 (2/0/2)
3 (0/2/1)
2 (0/1/1)
2 (1/0/1)
5 (2/0/3)
2 (1/0/1)
Ace!cAcid= CytC-MT
MCD1-MT = CytC-MT
CytC-MT = CytC-CYT
CytC-CYT =YCA1
!SRO7 + Salt =YCA1
MEC1=YCA1
MT-Frag =YCA1
ROS-CYT =YCA1
RedAc!nDyn =YCA1
ESP1 = ROS-CYT
SOD1 = ROS-CYT
2 NDI1 = ROS-CYT
Salt = ROS-CYT
RAS2 = ROS-CYT
RedAc!nDyn = ROS-CYT
H2O2 + !EMC4 = ROS-CYT
H2O2 + !SVF1 = ROS-CYT
Ace!cAcid + !SVF1 = ROS-CYT
!SOD2 + NDI1 = ROS-MT
MT-ALT = MT-FRAG
!FIS1 + MDV1 = MT-Frag
H2O2 + !DRE2/TAH18 = MT-Frag
!SNO1 = apoptosis
!FYV10 = apoptosis
YCA1 + !BIR1 = apoptosis
ROS-CYT = Apoptosis
DNA-Frag = apoptosis
!STM1-NUC = DNA-Frag
!POR1-2 + H2O2 = apoptosis
!POR1-2 + Ace!cAcid = apoptosis
CPR3 = apoptosis
RedAc!nDyn = RAS2
TOR1 = RAS2
RAS2 = adenylatecyclase
Stress = AdenylateCyclase
AdenylateCyclase = CAMP
RAS2 = PKA
TOR1 = CAMP
PKA=MT-ALT
RAS2 = MT–ALT
MMI1 = MT-ALT
Adozelesin = CDC6
CDC6 = PROTOSOM
STM1-CYT + !Protosom = STM1-NUC
Heat = NMA111-CYT
NMA111-CYT = NMA111-NUC
!NMA111-NUC = BIR1
Ma!ng = MAPK
MAPK = STE20-CYT
STE20-CYT = STE20-NUC
Abnormal Telomer = MEC1
Stress = RPD3
HOS3 = H2B
RPD3 = H2B
NUC1-NUC = H2B
STE20-NUC = H2B
AIF1 -NUC = H2B
H2O2 = HOS3
ESP1 = MCD1-NUC
H2O2 = ESP1
H2O2 = NUC1-MT
NUC1-MT = KAP123
KAP123 = NUC1-NUC
AIF1-MT = AIF1-NUC
Tat-D = DNA-Frag
Mg2+=Tat-D
=Ace!cAcid
= Heat
=H2O2
=Adozelesin
=MG2
=CU2
=Salt
=Ma!ng
=Stress
Apoptosis=
Cu2 = CPR3
Heat = PKA
PKA = MSN2-4
MSN2-4 = SDP1
Heat = SLT2
!SDP1 = SLT2
SLT2 = RLM1
RLM1 = SLT2
RLM1 = PTP2
!PTP2 = SLT2
Stress = HOG1
Hog1 = RLM1
!PTP2 = HOG1
!PTP3 = HOG1
HOG1-Dep = PTP3
!Stress = SLT2
!SLT2 = DesCyclinCCDK8
DesCyclinCCCDK8 = ROS-CYT
HK = SVF1
HK = DRE2/TAH18
HK = EMC4
HK = POR1-2
HK = SNO1
HK=FVY10
HK = SRO7
HK = STM1-CYT
HK = MMI1
HK = NDI1
CDC48 = CytC-CYT
2 CDC48 = ROS-CYT
HK = CDC48
HK = FIS1
HK = MDV1
HK = MCD1-MT
HK = AIF1-MT
HOG1 = HOG1-Dep
HK = SOD2
Heat = SOD1
Stress =TOR1
FIGURE 9 | Dependency Matrix forYeast Apoptosis Network. In the
dependency matrix each element mij represent the relation between an
affecting and an affected species. Former is specified at the bottom of each
column and later is shown at the beginning of each row. At intersection of i th
column and j th row there are three possibilities. A yellow box indicates
species iis an ambivalent factor meaning both activating and inhibiting path
exist from species i to species j. Similarly a dark green and or light green cell
shows a total and a non-total activator respectively. Another possibility is
having dark or light red indicates species i is a total inhibitor or non-total
inhibitor of species j. whenever there is no path from spices i to species j
the intersection cell is field by black. The hyper graph underlying the network
is a directed graph and consequently the dependency matrix is
non-symmetric. (Color coding: light and dark green – complete and
incomplete activation, dark and light red – complete and incomplete
inhibitor, yellow indicates an ambivalent factor and black indicates that there
is no dependency between two species).
COMPUTATION OF LOGICAL STEADY STATES
Identification of Logical Steady States (LSSs) in a Boolean net-
work is an important task as they comprise the states in which a
gene-regulatory network resides most of the time. Strong biolog-
ical implication can be carried out by LSSs of the network. LSSs
can even be linked to phenotype (Kauffman, 1969). CNA (Klamt
et al., 2006) is used to calculate all possible LLSs based on specified
initial value for each gene and signal flow in network. LSSs are used
to evaluate the network behavior under perturbation and changes
in network structure.
IDENTIFYING NETWORK WIDE DEPENDENCY
Considering a pair of nodes (a, b) dependency is defined as the
influence of node a on node b and vice versa, node a influences
node b as follow: a is a total activator of b if there is an activat-
ing path between a and b, a is a total inhibitor of b is there is
an inhibition path from a to b or a is an ambivalent effecter of
b meaning that there is an intermediate node which is involved
in a negative feedback loop. Using CNA the interaction matrix
and dependency matrix were drawn from the network (Figures 8
and 9).
CONVERSION OF DISCRETE TO CONTINUOUS MODEL USING SQUAD
In order to convert the schematic network into Boolean model
we used SQUAD (Mendoza and Xenarios, 2006), a user friendly
graphical software which is suitable for modeling signaling net-
work where kinetic reactions are not available. Simulation in
SQUAD consists of three steps: (1) the network is first loaded
from the SBML file. Components of the network are presented as
nodes and value of each node represents the state of that node.
SQUAD converts the network to discrete dynamic model. Using
Boolean algorithms all steady sates in network are calculated. (2)
network is converted to a continues dynamic model generating
sets of ordinary differential equations (ODEs) and steady states
achieved in pervious step. (3) SQUAD allows perturbation in order
to understand the role of each node within the network.
Using Reduced Order Binary Decision Diagram (ROBDD), it
is possible to calculate steady states (Di Cara et al., 2007). ROBDD
is a memory efficient data structure which is widely used in elec-
tronic field and has been proven to work for large binary networks.
Moreover, ROBDD computes steady states for large networks
(n > 50) in matter of seconds.Another advantage of this algorithm
is its ability to identify the cyclic steady states. These oscillating
www.frontiersin.org
December 2012 | Volume 3 | Article 446 | 15
Kazemzadeh et al.
Boolean model of yeast apoptosis
states are reachable when system identifies a cyclic pattern instead
of one single state.
In the first step, schematic network is supplied to SQUAD
resulting in a discrete network, generating a set of either cyclic
or single steady states. These steady states are then used to convert
discrete model to continues. Given all calculated steady states or
cycles we examined all possible outcomes for each input. Depend-
ing on the desire form of output,discrete to continuous conversion
can be carried out either as a complete or progressive mode. Our
simulations are performed in the complete mode since cell under-
going apoptosis should die after certain time and it is expected to
maintain a constant level of apoptosis or survival (in case when
apoptosis is not activated) at the end of each run. As an opposite
to complete mode, the progressive model allows the user to stop
the simulation at any time even before reaching the steady states.
CONVERSION OF DISCRETE TO CONTINUOUS MODEL USING ODEFY
Reconstructing Boolean model of yeast apoptosis from qualitative
knowledge never gives details about concentration of molecules
in different time points. For this purpose the discrete Boolean
model is transformed to continuous model using Odefy. Odefy
uses the multivariate polynomial interpolation in order to trans-
form the logical rules into sets of ODEs. Yeast apoptosis Boolean
model is converted to continuous model using Hill Cube and nor-
malized Hill Cube where the Hill function is normalized to the
unit interval. Behavior of biochemical reactions can be seen as
a sigmoid Hill function represented as f (¯¯x) = ¯¯xn/(¯¯xn + kn).
Where, n is a Hill coefficient and determines the slope of the
curve and is a measurement of cooperativity of the interactions,
and parameter k corresponds to values 1 and 0 in the Boolean
model in the following manner: threshold value above given k in
Boolean model is considered as 1 (on state) and below as 0 (off
state).
ACKNOWLEDGMENTS
We would like to thank Dr. Andrea Molt for initial discussions
and the Knut and Alice Wallenberg Foundation, the Chalmers
Foundation, and the FP7 EU Project SYSINBIO for funding.
REFERENCES
Aerts,A. M., Zabrocki, P., François, I. E.,
Carmona-Gutierrez, D., Govaert, G.,
Mao, C., et al. (2008). Ydc1p cerami-
dase triggers organelle fragmenta-
tion, apoptosis and accelerated age-
ing in yeast. Cell. Mol. Life Sci. 65,
1933–1942.
Ahn, S. H., Diaz, R. L., Grunstein, M.,
and Allis, C. D. (2006). Histone H2B
deacetylation at lysine 11 is required
for yeast apoptosis induced by phos-
phorylationof H2Batserine10.Mol.
Cell 24, 211–220.
Albertyn,
J.,
and
Hohmann,
S.
(1994).
Characterization
of
the
osmotic-stress response in Saccha-
romyces cerevisiae: osmotic stress
and
glucose
repression
regulate
glycerol-3-phosphate
dehydroge-
nase independently. Curr. Genet. 25,
12–18.
Almeida, B., Silva, A., Mesquita, A.,
Sampaio-Marques, B., Rodrigues, F.,
and Ludovico, P. (2008). Drug-
induced apoptosis in yeast. Biochim.
Biophys. Acta 7, 1436–1448.
Blanchard, F., Rusiniak, M. E., Sharma,
K., Sun, X., Todorov, I., Castel-
lano, M. M., et al. (2002). Targeted
destruction of DNA replication pro-
tein Cdc6 by cell death pathways in
mammals and yeast. Mol. Biol. Cell
13, 1536–1549.
Braun, R. J., and Zischka, H. (2008).
Mechanisms of Cdc48/VCP-medi-
ated cell death-from yeast apoptosis
to human disease. Biochim. Biophys.
Acta 1783, 1418–1435.
Brewster, J. L., de Valoir, T., Dwyer,
N. D, Winter, E., and Gustin, M.
C. (1993). An osmosensing signal
transduction pathway in yeast. Sci-
ence 259, 1760–1763.
Buttner, S., Eisenberg, T., Carmona-
Gutierrez, D., Ruli, D., Knauer,
H., Ruckenstuhl, C., et al. (2007).
Endonuclease G regulates budding
yeast life and death. Mol. Cell 25,
233–246.
Carmona-Gutierrez, D., Eisenberg, T.,
Büttner, S., Meisinger, C., Kroemer,
G.,and Madeo,F. (2010a).Apoptosis
in yeast: triggers, pathways, subrou-
tines. Cell. Death Differ. 17,763–773.
Carmona-Gutierrez, D., Ruckenstuhl,
C., Bauer, M. A., Eisenberg, T.,
Buttner, S., and Madeo, F. (2010b).
Cell death in yeast: growing applica-
tions of a dying buddy. Cell Death
Differ. 17, 733–734.
Cherry, J. M., Hong, E. L., Amund-
sen, C., Balakrishnan, R., Binkley,
G., Chan, E. T., et al. (2011). Sac-
charomyces genome database: the
genomics resource of budding yeast.
Nucleic Acids Res. 40, D700–D705.
[Database issue]
Datta, S. R., Dudek, H., Tao, X., Mas-
ters, S., Fu, H., Gotoh, Y., et al.
(1997).Akt phosphorylation of BAD
couples survival signals to the cell-
intrinsic death machinery. Cell 91,
231–241.
Davidich,
M.,
and
Bornholdt,
S.
(2008). Boolean network model
predicts cell cycle sequence of fis-
sion yeast. PLoS ONE
3:e1672.
doi:10.1371/journal.pone.0001672
de Nadal, E., Zapater, M., Alepuz, P.
M., Sumoy, L., Mas, G., and Posas,
P. (2004). The MAPK Hog1 recruits
Rpd3 histone deacetylase to activate
osmoresponsive genes. Nature 427,
370–374.
Di Cara, A., Garg, A., De Micheli,
G., Xenarios, I., and
Mendoza,
L.
(2007).
Dynamic
simulation
of
regulatory
networks
using
SQUAD. BMC Bioinformatics 8:462.
doi:10.1186/1471-2105-8-462
Drexler, H. C. A. (1997). Activation of
the cell death program by inhibition
of proteasome function. Proc. Natl.
Acad. Sci. U.S.A. 94, 855–860.
Eisenberg, T., Buttner, S., Kroemer, G.,
and Madeo, F. (2007). The mito-
chondrial pathway in yeast apopto-
sis. Apoptosis 12, 1011–1023.
Fahrenkrog, B., Sauder, U., and Aebi,
U. (2004). The S. cerevisiae HtrA-
like protein Nma111p is a nuclear
serine protease that mediates yeast
apoptosis. J. Cell Sci. 117, 115–126.
Falasca, M. (2010). PI3K/Akt signalling
pathway specific inhibitors: a novel
strategy to sensitize cancer cells to
anti-cancer drugs. Curr. Pharm. Des.
16, 1410–1416.
Fauré, A., Naldi, A., Chaouiya, C., and
Thieffry, D. (2006). Dynamic analy-
sis of a generic Boolean model for
the control of the mammalian cell
cycle. Bioinformatics 22, 124–131.
Ferrigno, P., Posas, F., Koepp, D., Saito,
H., and Silver, P. A. (1998). Reg-
ulated nucleo/cytoplasmic exchange
of
HOG1
MAPK
requires
the
importin beta homologs NMD5 and
XPO1. EMBO J. 17, 5606–5614.
Frohlich, K. U., Fussi, H., and Ruck-
enstuhl, C. (2007). Yeast apoptosis-
from genes to pathways. Semin. Can-
cer Biol. 17, 112–121.
Funahashi, A., Tanimura, N., Mat-
suoka, Y., Yosinaga, N., and Kitano,
H. (2003). CellDesigner: a process
diagram editor for gene-regulatory
and biochemical networks. BIOSIL-
ICO 1, 159–162.
Garbarino, J., Padamsee, M., Wilcox,
L., Oelkers, P. M., D’ Ambrosio,
D., Ruggles, K., et al. (2009). Sterol
and diacylglycerol acyltransferase
deficiency
triggers
fatty
acid-
mediated cell death. J. Biol. Chem.
284, 30994–31005.
Greenhalf, W., Stephan, C., and Chaud-
huri, B. (1996). Role of
mito-
chondria and C-terminal membrane
anchor of Bcl-2 in Bax induced
growth arrest and mortality in Sac-
charomycescerevisiae.FEBSLett.380,
169–175.
Gupta, S., Bisht, S. S., Kukreti, R., Jain,
S., and Brahmachari, S. K. (2007).
Boolean network analysis of neu-
rotransmitter signaling pathway. J.
Theor. Biol. 244, 463–469.
Hahn, J. S., and Thiele, D. J. (2002).
Regulation of the Saccharomyces
cerevisiae Slt2 kinase pathway by
the stress-inducible Sdp1 dual speci-
ficity phosphatase. J. Biol. Chem.
277, 21278–21284.
Handorf, T., and Klipp, E. (2012).
Modeling
mechanistic
biological
networks:
an
advanced
Boolean
approach.
Bioinformatics
28,
557–563.
Hauptmann,
P.,
Riel,
C.,
Kunz-
Schughart, L. A., Fröhlich, K. U.,
Madeo, F., and Lehle, L. (2006).
Defects in N-glycosylation induce
apoptosis in yeast. Mol. Microbiol.
59, 765–778.
Hers, I., Vincent, E. E., and Tavaré, J. M.
(2011). Akt signalling in health and
disease. Cell. Signal. 23, 1515–1527.
Hirabayashi, M., Inoue, K., Tanaka, K.,
Nakadate, K., Ohsawa, Y., Kamei, Y.,
et al. (2001). VCP/p97 in abnor-
mal protein aggregates, cytoplasmic
vacuoles, and cell death, phenotypes
relevant to neurodegeneration. Cell
Death Differ. 8, 977–984.
Frontiers in Physiology | Systems Biology
December 2012 | Volume 3 | Article 446 | 16
Kazemzadeh et al.
Boolean model of yeast apoptosis
Hohmann, S. (2002). Osmotic stress
signaling and osmoadaptation in
yeast. Microbiol. Mol. Biol. Rev. 66,
300–372.
Hohmann, S. (2009). Control of high
osmolarity signalling in the yeast
Saccharomyces cerevisiae. FEBS Lett.
583, 4025–4029.
Hucka, M., Finney, A., Sauro, H.
M.,
Bolouri,
H.,
Doyle,
J.
C.,
and Kitano, H. (2003). The sys-
tems
biology
markup
language
(SBML): a medium for representa-
tion and exchange of biochemical
network models. Bioinformatics 19,
524–531.
Imamuraab,
S.,
Ojimaa,
N.,
and
Yamashitaa,
M.
(2003).
Cold-
inducible expression of the cell
division cycle gene CDC48 and
its promotion of cell proliferation
during cold acclimation in zebrafish
cells. FEBS Lett. 549, 14–20.
Ishigaki, S., Hishikawa, N., Niwa, J.,
Iemura, S., Natsume, T., Hori, S., et
al. (2004). Physical and functional
interaction
between
dorfin
and
valosin-containing
protein
that
are
colocalized
in
ubiquitylated
inclusions
in
neurodegenerative
disorders.
J.
Biol.
Chem.
279,
51376–51385.
Janes, K. A.,Albeck, J. G., Sorger, S. G. P.
K., Lauffenburger, D. A., and Yaffe,
M. B. (2005). A systems model of
signaling identifies a molecular basis
set for cytokine-induced apoptosis.
Science 310, 1646–1653.
Kanehisa, M., and Goto, S. (2000).
KEGG: kyoto encyclopedia of genes
and genomes. Nucleic Acids Res. 28,
27–30.
Kataoka, T., Powers, S., McGill, C.,
Fasano, O., Strathern, J., Broach, J.,
et al. (1984). Genetic analysis of
yeast RAS1 and RAS2 genes. Cell 37,
437–445.
Kauffman, S. A. (1969). Metabolic sta-
bility and epigenesis in randomly
constructed genetic nets. J. Theor.
Biol. 22, 437–467.
Kharbanda, S., Pandey, P., Schofield, L.,
Israels, S., Roncinske, R., Yoshida,
K., et al. (1997). Role for Bcl-xL as
an inhibitor of cytosolic cytochrome
C accumulation in DNA damage-
induced apoptosis. Proc. Natl. Acad.
Sci. U.S.A. 13, 6939–6942.
Khoury,C. M.,Yang,Z.,Li,X.Y.,Vignali,
M., Fields, S., and Greenwood, M. T.
(2008). A TSC22-like motif defines
a novel antiapoptotic protein family.
FEMS Yeast Res. 8, 540–563.
Klamt, S., Saez-Rodriguez, J., Lindquist,
J. A., Simeoni, L., and Gilles, E.
D. (2006). A methodology for the
structural and functional analy-
sis
of
signaling
and
regulatory
networks. BMC Bioinformatics 7:56.
doi:10.1186/1471-2105-7-56
Krasley, E., Cooper, K. F., Mallory, M. J.,
Dunbrack, R., and Strich, R. (2006).
Regulation of the oxidative stress
response through Slt2p-dependent
destruction of cyclin C in Sac-
charomyces cerevisiae. Genetics 172,
1477–1486.
Krumsiek, J., Pölsterl, S., Wittmann,
D. M., and Theis, F. J. (2010).
Odefy – from discrete to continuous
models. BMC Bioinformatics 11:233.
doi:10.1186/1471-2105-11-233
Li, C., Donizelli, M., Rodriguez, N.,
Dharuri, H., Endler, L., Chelliah, V.,
et al. (2010). BioModels Database:
an enhanced, curated and annotated
resource for published quantitative
kinetic models. BMC Syst. Biol. 4:92.
doi:10.1186/1752-0509-4-92
Li, W., Sun, L., Liang, Q., Wang,
J., Mo, W., and Zhou, B. (2006).
Yeast AMID homologue Ndi1p dis-
plays respiration-restricted apop-
totic activity and is involved in
chronological aging. Mol. Biol. Cell
17, 1802–1811.
Liang, Q., and Zhou, B. (2007). Copper
andmanganeseinduceyeastapopto-
sis via different pathways. Mol. Biol.
Cell 18, 4741–4749.
Ligr, M., Velten, I., Fröhlich, E., Madeo,
F., Ledig, M., Fröhlich, K. U., et al.
(2001). The proteasomal substrate
Stm1 participates in apoptosis-like
cell death in yeast. Mol. Biol. Cell 12,
2422–2432.
Low, C. P., Shui, G., Liew, L. P., But-
tner, S., Madeo, F., Dawes, I. W., et
al. (2008). Caspase-dependent and
-independent lipotoxic cell-death
pathways in fission yeast. J. Cell. Sci.
15, 2671–2684.
Ludovico, P., Rodrigues, F., Almeida,
A., Silva, M. T., Barrientos, A., and
Côrte-Real, M. (2002). Cytochrome
c
release
and
mitochondria
involvement
in
programmed
cell death induced by acetic acid in
Saccharomyces cerevisiae. Mol. Biol.
Cell 13, 2598–2606.
Ludovico, P., Sousa, M. J., Silva, M. T.,
Leao, C., and Côrte-Real, M. (2001).
Saccharomyces cerevisiae commits
to a programmed cell death process
in response to acetic acid. Microbiol-
ogy 147, 2409–2415.
Madeo,
F.,
Carmona-Gutierrez,
D.,
Ring, J., Buttner, S., Eisenberg,
T.,
and
Kroemer,
G.
(2009).
Caspase-dependent
and
caspase-
independent cell death pathways
in yeast. Biochem. Biophys. Res.
Commun. 382, 227–231.
Madeo, F., Fröhlich, E., and Fröhlich,
K. U. (1997). A yeast mutant show-
ing diagnostic markers of early and
late apoptosis. J. Cell Biol. 139,
729–734.
Madeo, F., Fröhlich, E., Ligr, M., Grey,
M., Sigrist, S. J., Wolf, D. H., et al.
(1999). Oxygen stress: a regulator of
apoptosis in yeast. J. Cell Biol. 145,
757–767.
Madeo, F., Herker, E., Maldener, C.,
Wissing,S.,Lächelt,S.,Herlan,M.,et
al.(2002).Acaspase-relatedprotease
regulatesapoptosisinyeast.Mol.Cell
9, 911–917.
Mai, Z., and Liu, H. (2009). Boolean
network-based analysis of the apop-
tosis network: irreversible apoptosis
and stable surviving. J. Theor. Biol.
259, 760–769.
Manon, S., Chaudhuri, B., and Guérin,
M. (1997). Release of cytochrome
c and decrease of cytochrome c
oxidase in Bax-expressing yeast cells,
and prevention of these effects by
coexpression of Bcl-xL. FEBS Lett.
415, 29–32.
Mazzoni, C., Herker, E., Palermo, V.,
Jungwirth, H., Eisenberg, T., Madeo,
F., et al. (2005). Yeast caspase 1
links messenger RNA stability to
apoptosis in yeast. EMBO Rep. 6,
1076–1081.
Mendoza, L., and Xenarios, I. (2006). A
method for the generation of stan-
dardized qualitative dynamical sys-
tems of regulatory networks. Theor.
Biol. Med. Model. 3, 13.
Mizuno, Y., Hori, S., Kakizuka, A.,
and Okamoto, K. (2003). Vacuole-
creating protein in neurodegenera-
tive diseases in humans. Neurosci.
Lett. 343, 77–80.
Nelson, L. D., Musso, M., and Van Dyke,
M. W. (2000). The yeast STM1 gene
encodes a purine motif triple helical
DNA-binding protein. J. Biol. Chem.
275, 5573–5581.
Owsianowski,
E.,
Walter,
D.,
and
Fahrenkrog, B. (2008). Negative
regulation of apoptosis in yeast.
Biochim.
Biophys.
Acta
1783,
1303–1310.
Pereira, C., Camougrand, N., Manon,
S., Sousa, M. J., and Côrte-Real, M.
(2007). ADP/ATP carrier is required
for mitochondrial outer membrane
permeabilization and cytochrome
c release in yeast apoptosis. Mol.
Microbiol. 66, 571–582.
Qiu, J., Yoon, J. H., and Shen, B. (2005).
Search for apoptotic nucleases in
yeast: role of Tat-D nuclease in
apoptotic DNA degradation. J. Biol.
Chem. 280, 15370–15379.
Ring, G., Khoury, C. M., Solar, A. J.,
Yang, Z., Mandato, C. A., and Green-
wood, M. T. (2008). Transmem-
brane protein 85 from both human
(TMEM85) and yeast (YGL231c)
inhibit hydrogen peroxide mediated
cell death in yeast. FEBS Lett. 582,
2637–2642.
Ruchaud, S., Carmena, M., and Earn-
shaw, W. C. (2007). Chromoso-
mal passengers: conducting cell divi-
sion. Nat. Rev. Mol. Cell Biol. 8,
798–812.
Sato, T., Hanada, M., Bodrug, S., Irie,
S., Iwama, N., Boise, L. H., et al.
(1994). Interactions among mem-
bers of the Bcl-2 protein family ana-
lyzed with a yeast two hybrid sys-
tem. Proc. Natl. Acad. Sci. U.S.A. 91,
9238–9242.
Schlatter,R.,Schmich,K.,Vizcarra,I. A.,
Scheurich, P., Sauter, T., Borner, C.,
et al. (2009). On/off and beyond –
a
boolean
model
of
apoptosis.
PLoS
Comput.
Biol.
5:e1000595.
doi:10.1371/journal.pcbi.1000595
Schmelzle,T.,Beck,T.,Martin,D.E.,and
Hall, M. N. (2004). Activation of the
RAS/cyclic AMP pathway suppresses
a TOR deficiency in yeast. Mol. Cell
Biol. 24, 338–351.
Song, C., Wang, Q., and Li, C. C.
(2003). ATPase activity of p97-
valosin-containing protein (VCP).
D2 mediate the major enzyme activ-
ity and D1 contributes in heat-
induced activity. J. Biol. Chem. 278,
3648–3655.
Stelling,J. (2004). Mathematical models
in microbial systems biology. Curr.
Opin. Microbiol. 7, 513–518.
Thomas, R., and D’Ari, R. (1990). Bio-
logical Feedback. Boca Raton: CRC
Press.
Uren, A.
G., Coulson, E.
J., and
Vaux, D. L. (1998). Conservation
of baculovirus inhibitor of apop-
tosis repeat proteins (BIRPs) in
viruses, nematodes, vertebrates and
yeasts. Trends Biochem. Sci. 23,
159–162.
VanWuytswinkel,O.,Reiser,V.,Siderius,
M., Kelders, M. C., Ammerer, G.,
Ruis, H., et al. (2000). Response of
Saccharomyces cerevisiae to severe
osmotic stress: evidence for a novel
activation mechanism of the HOG
MAP kinase pathway. Mol. Micro-
biol. 37, 382–397.
Vander Heiden, M. G., Choy, J. S., Van-
derWeele, D. J., Brace, J. L., Harris,
M. H.,Bauer, D. E.,et al. (2002). Bcl-
x(L) complements Saccharomyces
cerevisiae genes that facilitate the
switch from glycolytic to oxida-
tive metabolism. J. Biol. Chem. 277,
44870–44876.
Vandermoere, F., El Yazidi-Belkoura,
I.,
Slomianny,
C.,
Demont,
Y.,
Bidaux, G., Adriaenssens, E., et al.
(2006). The valosin-containing pro-
tein (VCP) is a target of Akt signal-
ing required for cell survival. J. Biol.
Chem. 20, 14307–14313.
www.frontiersin.org
December 2012 | Volume 3 | Article 446 | 17
Research Topic:
From structural to molecular systems biology: experimental and
computational approaches to unravel mechanisms of kinase activity
regulation in cancer and neurodegeneration
Topic Editor:
Matteo Barberis,
Humboldt University Berlin, Germany;
Max Planck Institute for Molecular Genetics, Berlin, Germany
Kazemzadeh et al.
Boolean model of yeast apoptosis
Vernis, L., Facca, C., Delagoutte, E.,
Soler, N., Chanet, R., Guiard, B.,
et al. (2009). A newly identified
essential complex, Dre2-Tah18, con-
trols mitochondria integrity and
cell
death
after
oxidative
stress
in
yeast.
PLoS
ONE
4:e4376.
doi:10.1371/journal.pone.0004376
Wadskog, I., Maldener, C., Proksch, A.,
Madeo, F., and Adler, L. (2004).Yeast
lacking
the
SRO7/SOP1-encoded
tumor suppressor homologue show
increasedsusceptibilitytoapoptosis-
like cell death on exposure to
NaCl stress. Mol. Biol. Cell 15,
1436–1444.
Walter, D., Wissing, S., Madeo, F.,
and Fahrenkrog, B. (2006). The
inhibitor-of-apoptosis protein Bir1p
protects against apoptosis in S.
cervisiae and is a substrate for the
yeast homologue of Omi/HtrA2. J.
Cell. Sci. 119, 1843–1851.
Wang, Q., Song, C., and Li, C. H.
(2004). Molecular perspective on
p97-VCP: progress in understand-
ing its structure and diverse bio-
logical function. J. Struct. Biol. 164,
44–57.
Warmka, J., Hanneman, J., Lee, J.,Amin,
D., and Ota, I. (2001). Ptc1, a type
2C Ser/Thr phosphatase, inactivates
the HOG pathway by dephosphory-
lating the mitogen-activated protein
kinase Hog1. Mol. Cell. Biol. 21,
51–60.
Weinberger, M., Ramachandran, L.,
Feng, L., Sharma, K., Sun, X., Mar-
chetti, M., et al. (2005). Apoptosis in
budding yeast caused by defects in
initiation of DNA replication. J. Cell
Sci. 118, 3543–3553.
Westfall,P. J.,Ballon,D. R.,and Thorner,
J. (2004). When the stress of your
environment makes you go HOG
wild. Science 306, 1511–1512.
Winderickx, J., Delay, C., De Vos,
A., Klinger, H., Pellens, K., Van-
helmont, T., et al. (2008). Pro-
tein folding diseases and neurode-
generation: lessons learned from
yeast. Biochim. Biophys. Acta 1783,
1381–1395.
Winkler, A., Arkind, C., Mattison, C.
P., Burkholder, A., Knoche, K., and
Ota, I. (2002). Heat stress activates
the yeast high-osmolarity glycerol
mitogen-activated
protein
kinase
pathway, and protein tyrosine phos-
phatases are essential under heat
stress. Eukaryot. Cell 1, 163–173.
Wissing,S.,Ludovico,P.,Herker,E.,But-
tner,S.,Engelhardt,S. M.,Decker,T.,
et al. (2004). An AIF orthologue reg-
ulates apoptosis in yeast. J. Cell Biol.
166, 969–974.
Wittmann, D. M., Krumsiek, J., Saez-
Rodriguez, J., Lauffenburger, D. A.,
Klamt, S., and Theis, F. J. (2009).
Transforming Boolean models to
continuous models: methodology
and application to T-cell recep-
tor signaling. BMC Syst. Biol. 3:98.
doi:10.1186/1752-0509-3-98
Wolkenhauer, O. (2002). Mathemati-
cal modelling in the post-genome
era: under-standing genome expres-
sion and regulation – a system
theoretic approach. BioSystems 65,
1–18.
Wood, D. R., Poullet, P., Wilson, B. A.,
Khalil, M., Tanaka, K., Cannon, J.
F., et al. (1994). Biochemical char-
acterization of yeast RAS2 mutants
reveals a new region of ras pro-
tein involved in the interaction with
GTPase-activating proteins. J. Biol.
Chem. 269, 5322–5327.
Yang, H., Ren, Q., and Zhang, Z. (2008).
Cleavage of Mcd1 by caspase-like
protease Esp1 promotes apoptosis in
budding yeast. Mol. Biol. Cell 19,
2127–2134.
Young, C., Mapes, J., Hanneman, J., Al-
Zarban, S., and Ota, I. (2002). Role
of Ptc2 type 2C Ser/Thr phosphatase
in yeast high-osmolarity glycerol
pathway inactivation. Eukaryot. Cell
1, 1032–1040.
Zhang, N. N., Dudgeon, D. D., Pali-
wal, S., Levchenko, A., Grote, E.,
and Cunningham, K. W. (2006).
Multiple signaling pathways regulate
yeast cell death during the response
to mating pheromones. Mol. Biol.
Cell 17, 3409–3422.
Conflict of Interest Statement: The
authors declare that the research was
conducted
in
the
absence
of
any
commercial or financial relationships
that could be construed as a potential
conflict of interest.
Received: 29 June 2012; accepted: 07
November 2012; published online: 10
December 2012.
Citation: Kazemzadeh L, Cvijovic M
and
Petranovic
D
(2012)
Boolean
model of yeast apoptosis as a tool
to study yeast and human apoptotic
regulations. Front. Physio. 3:446. doi:
10.3389/fphys.2012.00446
This article was submitted to Frontiers in
Systems Biology, a specialty of Frontiers
in Physiology.
Copyright © 2012 Kazemzadeh, Cvi-
jovic and Petranovic. This is an open-
access article distributed under the terms
of the Creative Commons Attribution
License, which permits use, distribution
and reproduction in other forums, pro-
vided the original authors and source
are credited and subject to any copy-
right notices concerning any third-party
graphics etc.
Frontiers in Physiology | Systems Biology
December 2012 | Volume 3 | Article 446 | 18
|
23233838
|
SDP1 = ( MSN2-4 )
CDC48 = ( HK )
NDI1 = ( HK )
MT-ALT = ( MMI1 ) OR ( RAS2 ) OR ( MT-Frag ) OR ( PKA )
MEC1 = ( AbnormalTelomer )
YCA1 = ( ( Salt ) AND NOT ( SRO7 ) ) OR ( MEC1 ) OR ( MT-Frag ) OR ( RedActinDyn ) OR ( ROS-CYT ) OR ( CytC-CYT )
DesCyclinCCDK8 = NOT ( ( SLT2 ) )
Apoptosis = ( ( POR1-2 AND ( ( ( NOT AceticAcid OR NOT BIR1 OR NOT H2O2 OR NOT FVY10 ) AND ( ( ( NOT SNO1 ) ) ) ) ) ) OR ( CU2 AND ( ( ( CPR3 ) ) OR ( ( NOT POR1-2 OR NOT AceticAcid OR NOT BIR1 OR NOT H2O2 ) AND ( ( ( NOT FVY10 AND NOT SNO1 ) ) ) ) ) ) OR ( CPR3 AND ( ( ( NOT POR1-2 OR NOT AceticAcid OR NOT BIR1 OR NOT H2O2 OR NOT FVY10 ) AND ( ( ( NOT SNO1 ) ) ) ) ) ) OR ( ROS-CYT AND ( ( ( NOT POR1-2 OR NOT AceticAcid OR NOT BIR1 OR NOT H2O2 OR NOT FVY10 OR NOT SNO1 ) ) OR ( ( POR1-2 AND AceticAcid AND BIR1 AND H2O2 ) ) ) ) OR ( ( YCA1 AND ( ( ( NOT POR1-2 OR NOT AceticAcid OR NOT BIR1 OR NOT H2O2 OR NOT FVY10 OR NOT SNO1 ) ) ) ) AND NOT ( BIR1 AND ( ( ( FVY10 AND SNO1 ) ) ) ) ) OR ( DNA-Frag AND ( ( ( NOT POR1-2 OR NOT AceticAcid OR NOT BIR1 OR NOT H2O2 OR NOT FVY10 OR NOT SNO1 ) ) OR ( ( POR1-2 AND AceticAcid AND BIR1 AND H2O2 AND FVY10 ) ) ) ) OR ( BIR1 AND ( ( ( NOT POR1-2 AND NOT AceticAcid AND NOT H2O2 AND NOT FVY10 AND NOT SNO1 ) ) ) ) OR ( FVY10 AND ( ( ( NOT POR1-2 OR NOT AceticAcid OR NOT BIR1 OR NOT H2O2 ) AND ( ( ( NOT SNO1 ) ) ) ) ) ) OR ( SNO1 AND ( ( ( H2O2 AND FVY10 ) AND ( ( ( NOT POR1-2 AND NOT DNA-Frag AND NOT ROS-CYT ) ) ) ) OR ( ( NOT POR1-2 OR NOT AceticAcid OR NOT BIR1 OR NOT H2O2 ) AND ( ( ( NOT FVY10 ) ) ) ) ) ) OR ( H2O2 AND ( ( ( NOT POR1-2 OR NOT AceticAcid OR NOT BIR1 OR NOT FVY10 ) AND ( ( ( NOT SNO1 ) ) ) ) ) ) OR ( AceticAcid AND ( ( ( FVY10 AND SNO1 ) AND ( ( ( NOT POR1-2 AND NOT YCA1 AND NOT DNA-Frag AND NOT ROS-CYT AND NOT BIR1 AND NOT H2O2 ) ) ) ) OR ( ( NOT POR1-2 OR NOT BIR1 OR NOT H2O2 OR NOT FVY10 ) AND ( ( ( NOT SNO1 ) ) ) ) OR ( ( POR1-2 AND BIR1 AND H2O2 ) AND ( ( ( NOT FVY10 OR NOT SNO1 ) ) ) ) OR ( ( BIR1 AND FVY10 AND SNO1 ) AND ( ( ( NOT POR1-2 AND NOT H2O2 ) ) ) ) ) ) ) OR NOT ( POR1-2 OR AceticAcid OR DNA-Frag OR YCA1 OR ROS-CYT OR BIR1 OR H2O2 OR CU2 OR FVY10 OR SNO1 OR CPR3 )
SVF1 = ( HK )
PTP3 = ( HOG1-Dep )
SOD2 = ( HK )
POR1-2 = ( HK )
BIR1 = NOT ( ( NMA111-NUC ) )
DRE2_TAH18 = ( HK )
STM1-NUC = ( ( STM1-CYT ) AND NOT ( Protosom ) )
RLM1 = ( HOG1 ) OR ( SLT2 )
SLT2 = ( ( RLM1 ) OR ( Heat ) OR ( PTP2 AND ( ( ( NOT SDP1 OR NOT Stress ) ) ) ) OR ( SDP1 AND ( ( ( NOT PTP2 OR NOT Stress ) ) ) ) OR ( Stress AND ( ( ( NOT SDP1 OR NOT PTP2 ) ) ) ) ) OR NOT ( SDP1 OR PTP2 OR Stress OR Heat OR RLM1 )
MCD1-MT = ( HK )
FIS1 = ( HK )
DNA-Frag = ( ( Tat-D ) ) OR NOT ( Tat-D OR STM1-NUC )
CAMP = ( AdenylateCyclase ) OR ( TOR1 )
KAP123 = ( NUC1-MT )
STE20-CYT = ( MAPK )
SOD1 = ( Heat )
HOG1 = ( ( Stress ) OR ( PTP3 AND ( ( ( NOT PTP2 ) ) ) ) OR ( PTP2 AND ( ( ( NOT PTP3 ) ) ) ) ) OR NOT ( PTP3 OR PTP2 OR Stress )
AdenylateCyclase = ( Stress ) OR ( RAS2 )
ROS-CYT = ( Salt ) OR ( CDC48 ) OR ( RAS2 ) OR ( DesCyclinCCDK8 ) OR ( NDI1 ) OR ( RedActinDyn ) OR ( ( ( H2O2 ) AND NOT ( SVF1 ) ) AND NOT ( EMC4 ) ) OR ( ( AceticAcid ) AND NOT ( SVF1 ) ) OR ( ESP1 ) OR ( SOD1 )
Tat-D = ( MG2 )
HOS3 = ( H2O2 )
FVY10 = ( HK )
MDV1 = ( HK )
NMA111-NUC = ( NMA111-CYT )
PTP2 = ( RLM1 )
CytC-MT = ( MCD1-MT ) OR ( AceticAcid )
EMC4 = ( HK )
H2B = ( RPD3 ) OR ( AIF1-NUC ) OR ( STE20-NUC ) OR ( HOS3 ) OR ( NUC1-NUC )
ROS-MT = ( NDI1 AND ( ( ( NOT SOD2 ) ) ) )
PKA = ( Heat ) OR ( RAS2 )
CytC-CYT = ( CDC48 ) OR ( CytC-MT )
MT-Frag = ( ( H2O2 ) AND NOT ( DRE2_TAH18 ) ) OR ( MT-ALT ) OR ( ( MDV1 ) AND NOT ( FIS1 ) )
MSN2-4 = ( PKA )
STM1-CYT = ( HK )
NUC1-NUC = ( KAP123 )
ESP1 = ( H2O2 )
RAS2 = ( TOR1 ) OR ( RedActinDyn )
MMI1 = ( HK )
HOG1-Dep = ( HOG1 )
NMA111-CYT = ( Heat )
NUC1-MT = ( H2O2 )
RPD3 = ( Stress )
AIF1-MT = ( HK )
TOR1 = ( Stress )
MCD1-NUC = ( ESP1 )
AIF1-NUC = ( AIF1-MT )
CDC6 = ( Adozelesin )
Protosom = ( CDC6 )
MAPK = ( Mating )
STE20-NUC = ( STE20-CYT )
SNO1 = ( HK )
SRO7 = ( HK )
|
Canalization and Control in Automata Networks: Body
Segmentation in Drosophila melanogaster
Manuel Marques-Pita1,2*, Luis M. Rocha1,2*
1 Instituto Gulbenkian de Cieˆncia, Oeiras, Portugal, 2 Indiana University, Bloomington, Indiana, United States of America
Abstract
We present schema redescription as a methodology to characterize canalization in automata networks used to model
biochemical regulation and signalling. In our formulation, canalization becomes synonymous with redundancy present in
the logic of automata. This results in straightforward measures to quantify canalization in an automaton (micro-level), which
is in turn integrated into a highly scalable framework to characterize the collective dynamics of large-scale automata
networks (macro-level). This way, our approach provides a method to link micro- to macro-level dynamics – a crux of
complexity. Several new results ensue from this methodology: uncovering of dynamical modularity (modules in the
dynamics rather than in the structure of networks), identification of minimal conditions and critical nodes to control the
convergence to attractors, simulation of dynamical behaviour from incomplete information about initial conditions, and
measures of macro-level canalization and robustness to perturbations. We exemplify our methodology with a well-known
model of the intra- and inter cellular genetic regulation of body segmentation in Drosophila melanogaster. We use this
model to show that our analysis does not contradict any previous findings. But we also obtain new knowledge about its
behaviour: a better understanding of the size of its wild-type attractor basin (larger than previously thought), the
identification of novel minimal conditions and critical nodes that control wild-type behaviour, and the resilience of these to
stochastic interventions. Our methodology is applicable to any complex network that can be modelled using automata, but
we focus on biochemical regulation and signalling, towards a better understanding of the (decentralized) control that
orchestrates cellular activity – with the ultimate goal of explaining how do cells and tissues ‘compute’.
Citation: Marques-Pita M, Rocha LM (2013) Canalization and Control in Automata Networks: Body Segmentation in Drosophila melanogaster. PLoS ONE 8(3):
e55946. doi:10.1371/journal.pone.0055946
Editor: Luı´s A. Nunes Amaral, Northwestern University, United States of America
Received September 10, 2012; Accepted January 3, 2013; Published March 8, 2013
Copyright: 2013 Marques-Pita, Rocha. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: Funding provided by Fundac¸a˜o para a Ciencia e a Tecnologia (Portugal) grant PTDC/EIA-CCO/114108/2009. The funders had no role in study design,
data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors declare that LMR is a PLOS ONE Editorial Board member and that this does not alter the authors’ adherence to all the PLOS
ONE policies on sharing data and materials.
* E-mail: marquesm@indiana.edu (MMP); rocha@indiana.edu (LMR)
Introduction and Background
The notion of canalization was proposed by Conrad Waddington
[1] to explain why, under genetic and environmental perturba-
tions, a wild-type phenotype is less variable in appearance than
most mutant phenotypes during development. Waddington’s
fundamental hypothesis was that the robustness of wild-type
phenotypes is the result of a buffering of the developmental process. This
led Waddington to develop the well-known concept of epigenetic
landscape [2,3], where cellular phenotypes are seen, metaphorically,
as marbles rolling down a sloped and ridged landscape as the result
of interactions amongst genes and epigenetic factors. The marbles
ultimately settle in one of the valleys, each corresponding to a
stable configuration that can be reached via the dynamics of the
interaction network. In this view, genetic and epigenetic pertur-
bations can only have a significant developmental effect if they
force the natural path of the marbles over the ridges of the
epigenetic landscape, thus making them settle in a different valley
or stable configuration.
Canalization, understood as the buffering of genetic and
epigenetic perturbations leading to the stability of phenotypic traits,
has re-emerged recently as a topic of interest in computational and
systems biology [4–10]. However, canalization is an emergent
phenomenon because we can consider the stability of a phenotypic
trait both at the micro-level of biochemical interactions, or at the
macro-level of phenotypic behaviour. The complex interaction
between micro- and macro-level thus makes canalization difficult to
study in biological organisms – but the field of complex systems has
led to progress in our understanding of this concept. For instance,
Conrad [3] provided a still-relevant treatment of evolvability [11] by
analysing the tradeoff between genetic (micro-level) instability and
phenotypic (macro-level) stability. This led to the concept of extra-
dimensional bypass, whereby most genetic perturbations are buffered
to allow the phenotype to be robust to most physiological
perturbations, but a few genetic perturbations (e.g. the addition of
novel genetic information) provide occasional instability necessary
for evolution. Conrad highlighted three (micro-level) features of the
organization of living systems that allows them to satisfy this
tradeoff: modularity (or compartmentalization), component redundancy,
and multiple weak interactions. The latter two features are both a form
of redundancy, the first considering the redundancy of components
and the second considering the redundancy of interactions or
linkages. Perhaps because micro-level redundancy has been posited
as one of the main mechanisms to obtain macro-level robustness,
the term canalization has also been used – especially in discrete
mathematics – to characterize redundant properties of automata
functions, particularly when used to model micro-level dynamical
PLOS ONE | www.plosone.org
1
March 2013 | Volume 8 | Issue 3 | e55946
interactions in models of genetic regulation and biochemical
signalling.
An automaton is typically defined as canalizing if there is at least
one state of at least one of its inputs that is sufficient to control the
automaton’s next state (henceforth transition), regardless of the
states of any other inputs [12]. Clearly, this widely used definition
refers
to
micro-level
characteristics
of
the
components
of
multivariate
discrete
dynamical
systems
such
as
automata
networks, and not to canalization as the emergent phenomenon
outlined above. Nonetheless, using this definition, it has been
shown that (1) canalizing functions are widespread in eukaryotic
gene-regulation dynamics [13]; (2) genetic regulatory networks
modelled with canalizing automata are always stable [14,15]; and
(3) realistic biological dynamics are naturally observed in networks
with scale-free connectivity that contain canalizing functions [16].
These observations suggest that the redundancy captured by this
micro-level definition of canalization is a mechanism used to
obtain stability and robustness at the macro-level of phenotypic
traits.
Since the proportion of such ‘strictly’ canalizing functions drops
abruptly with the number of inputs (k) [17], it was at first assumed
that (micro-level) canalization does not play a prominent role in
stabilizing
complex
dynamics
of
gene
regulatory
networks.
However, when the concept of canalization is extended to include
partially canalizing functions, where subsets of inputs can control the
automaton’s transition, the proportion of available canalizing
automata increases dramatically even for automata with many
inputs [18]. Furthermore, partial canalization has been shown to
contribute to network stability, without a detrimental effect on
‘evolvability’ [18]. Reichhardt and Bassler, point out that, even
though strictly canalizing functions clearly contribute to network
stability, they can also have a detrimental effect on the ability of
networks to adapt to changing conditions [18] – echoing Conrad’s
tradeoff outlined above. This led them to consider the wider class
of partially canalizing functions that confer stable network
dynamics, while improving adaptability. A function of this class
may ignore one or more of its inputs given the states of others, but
is not required to have a single canalizing input. For example, if a
particular input is on, the states of the remaining inputs are
irrelevant, but if that same input is off, then the state of a subset of
its other inputs is required to determine the function’s transition.
In scenarios where two or more inputs are needed to determine
the transition, the needed inputs are said to be collectively canalizing.
Reichhardt and Bassler [18] have shown that the more general
class of partially canalizing functions dominates the space of
Boolean functions for any number of inputs k. Indeed, for any
value of k, there are only two non-canalizing functions that always
depend on the states of all inputs. Other classes of canalizing
functions have been considered, such as nested canalizing functions
[14], Post classes [19] and chain functions [20]. All these classes of
functions characterize situations of input redundancy in automata.
In other words, micro-level canalization is understood as a form of
redundancy, whereby a subset of input states is sufficient to
guarantee transition, and therefore its complement subset of input
states is redundant. This does not mean that redundancy is
necessarily the sole – or even most basic – mechanism to explain
canalization at the macro-level. But the evidence we reviewed
above, at the very least, strongly suggests that micro-level
redundancy is a key mechanism to achieve macro-level canaliza-
tion. Other mechanisms are surely at play, such as the topological
properties of the networks of micro-level interactions. Certainly,
modularity, as suggested by Conrad, plays a role in the robustness
of complex systems and has rightly received much attention
recently [21]. While we show below that some types of modularity
can derive from micro-level redundancy, other mechanisms to
achieve modularity are well-known [21].
Here, we explore partial canalization, as proposed by Reich-
hardt and Bassler [18], to uncover the loci of control in complex
automata networks, particularly those used as models of genetic
regulation and signalling. Moreover, we extend this notion to
consider not only (micro-level) canalization in terms of input
redundancy, but also in terms of input-permutation redundancy to
account for the situations in which swapping the states of (a subset)
of inputs has no effect on an automaton’s transition. From this
point forward, when we use the term canalization we mean it in the
micro-level sense used in the (discrete dynamical systems) literature
to characterize redundancy in automata functions. Nonetheless,
we show that the quantification of such micro-level redundancy
uncovers important details of macro-level dynamics in automata
networks used to model biochemical regulation. This allows us to
better study how robustness and control of phenotypic traits arises
in such systems, thus moving us towards understanding canaliza-
tion in the wider sense proposed by Waddington. Before
describing our methodology, we introduce necessary concepts
and notations pertaining to Boolean automata and networks, as
well as the segment polarity gene-regulation network in Drosophila
melanogaster, an automata model we use to exemplify our approach.
Boolean Networks
This type of discrete dynamical system was introduced to build
qualitative models of genetic regulation, very amenable to large-
scale statistical analysis [22] – see [23] for comprehensive review.
A Boolean automaton is a binary variable, x[f0,1g, where state 0 is
interpreted as false (off or unexpressed), and state 1 as true (on or
expressed). The states of x are updated in discrete time-steps, t,
according
to
a
Boolean
state-transition
function
of
k
inputs:
xtz1~f it
1,:::,it
k
. Therefore f : f0,1gk?f0,1g. Such a function
can be defined by a Boolean logic formula or by a look-up (truth) table
(LUT)
with
2k
entries.
An
example
of
the
former
is
xtz1~f (x,y,z)~xt ^ (yt _ zt), or its more convenient shorthand
representation f ~x ^ (y _ z), which is a Boolean function of k~3
input binary variables x,y,z, possibly the states of other automata;
^, _ and : denote logical conjunction, disjunction, and negation
respectively. The LUT for this function is shown in Figure 1. Each
LUT entry of an automaton x, fa, is defined by (1) a specific
condition, which is a conjunction of k inputs represented as a unique
k-tuple of input-variable (Boolean) states, and (2) the automaton’s
next state (transition) xtz1, given the condition (see Figure 1). We
denote the entire state transition function of an automaton x in its
LUT representation as F:ffa : a~1,:::,2kg.
A Boolean Network (BN) is a graph B:(X,E), where X is a set of
n Boolean automata nodes xi[X,i~1,:::,n, and E is a set of
directed edges eji[E : xi,xj[X. If eji[E, it means that automaton
xj
is
an
input
to
automaton
xi,
as
computed
by
Fi.
Xi~fxj[X : eji[Eg denotes the set of input automata of xi. Its
cardinality, ki~jXij, is the in-degree of node xi, which determines
the size of its LUT, jFij~2ki. We refer to each entry of Fi as
fi:a, a~1:::2ki. The input nodes of B are nodes whose state does not
depend on the states of other nodes in B. The state of output nodes is
determined by the states of other nodes in the network, but they
are not an input to any other node. Finally, the state of inner nodes
depends on the state of other nodes and affect the state of other
nodes in B. At any given time t, B is in a specific configuration of
node states, xt~Sx1,x2,:::,xnT. We use the terms state for
individual automata (x) and configuration (x) for the collection of
states of the set of automata of B, i.e. the collective network state.
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
2
March 2013 | Volume 8 | Issue 3 | e55946
Starting from an initial configuration, x0, a BN updates its
nodes with a synchronous or asynchronous policy. The dynamics of B is
thus defined by the temporal sequence of configurations that
ensue, and there are 2n possible configurations. The transitions
between configurations can be represented as a state-transition graph,
STG, where each vertex is a configuration, and each directed edge
denotes a transition from xt to xtz1. The STG of B thus encodes
the network’s entire dynamical landscape. Under synchronous
updating, configurations that repeat, such that xtzm~xt, are
known as attractors; fixed point when m~1, and limit cycle – with
period m – when mw1, respectively. The disconnected subgraphs
of a STG leading to an attractor are known as basins of attraction. In
contrast, under asynchronous updating, there are alternative
configuration transitions that depend on the order in which nodes
update their state. Therefore, the same initial configuration can
converge to distinct attractors with some probability [24,25]. A BN
B has a finite number b of attractors; each denoted by
Ai : i~1,:::,b. When the updating scheme is known, every
configuration x is in the basin of attraction of some specific
attractor Ai. That is, the dynamic trajectory of x converges to Ai.
We denote such a dynamical trajectory by s(x)
Ai. If the
updating scheme is stochastic, the relationship between configu-
rations and attractors can be specified as the conditional
probability P(Aijx).
The Segment Polarity Network
The methodology introduced in this paper will be exemplified
using the well-studied Boolean model of the segment polarity
network in Drosophila melanogaster [26]. During the early ontogenesis
of the fruit fly, like in every arthropod’s development, there is body
segmentation [27,28]. The specification of adult cell types in each
of these segments is controlled by a hierarchy of around forty
genes. While the effect of most of the genes in the hierarchy is only
transient, a subset of segment polarity genes remains expressed during
the life of the fruit fly [29]. The dynamics of the segment polarity
network was originally modelled using a system of non-linear
ordinary differential equations (ODEs) [30,31]. This model
suggested that the regulatory network of segment polarity genes
is a module largely controlled by external inputs that is robust to
changes to its internal kinetic parameters. On that basis, Albert
and Othmer later proposed a simpler discrete BN model of the
dynamics of the segment polarity network (henceforth SPN) [26]. This
was the first Boolean model of gene regulation capable of
predicting the steady state patterns experimentally observed in
wild-type and mutant embryonic development with significant
accuracy, and has thus become the quintessential example of the
power of the logical approach to modelling of biochemical
regulation from qualitative data in the literature. Modelling with
ODEs, in contrast, is hindered by the need of substantial
quantitative data for parameter estimation [32–37].
The SPN model comprises fifteen nodes that represent intra-
cellular chemical species and the genes engrailed (en); wingless (wg);
hedgehog (hh); patched (ptc) and cubitus interruptus (ci) [29–31]. These
genes encode a number of proteins such as the transcription
factors Engrailed (EN), Cubitus Interruptus (CI), CI Activator
(CIA), and CI repressor (CIR); the secreted proteins Wingless
(WG) and Hedgehog (HH); and the transmembrane protein
Patched (PTC). Other proteins included in the SPN model are
Sloppy-Paired (SLP) – the state of which is previously determined
by the pair-rule gene family that stabilizes its expression before the
segment polarity genes – as well as Smoothened (SMO) and the
PH complex that forms when HH from neighbouring cells binds
to PTC. Figure 2 shows the topology and Table 1 lists the logical
rules of the nodes in every cell of the SPN. This model consists of a
spatial arrangement of four interconnected cells, a parasegment.
While the regulatory interactions within each cell are governed by
the same network, inter-cellular signalling affects neighbouring
cells. That is, regulatory interactions in a given cell depend on the
states of WG, hh and HH in adjacent cells. Therefore, six
additional (inter-cellular) ‘spatial signals’ are included: hhi+1,
HHi+1 and WGi+1, where i~1,:::,4 is the cell index in the four-
cell parasegment. In a parasegment, the cell with index i~1
corresponds to its anterior cell and the cell with index i~4 to its
posterior
cell
(see
Figure
3).
In
simulations,
the
four-cell
parasegments assume periodic boundary conditions (i.e. anterior
and posterior cells are adjacent to each other). Since each
parasegment has 4|15~60 nodes, four of which are in a fixed
state (SLP), there are 256 possible configurations – a dynamical
landscape too large for exhaustive analysis. Even though the
original model was not fully synchronous because PH and SMO
were updated instantaneously at time t, rather than at tz1, here
we use the fully equivalent, synchronous version. Specifically, since
PH is an output node, synchronizing its transition with the
remaining nodes at tz1 does not impact the model’s dynamics.
The state of SMO influences the updating of CIA and CIR; but
since the update of SMO is instantaneous, we can include its state-
transition function in the state-transition functions of CIA and
CIR (which update at tz1) with no change in the dynamics of the
model as described in [38].
The initial configuration (IC) of the SPN, depicted in Figure 3,
and which leads to the wild-type expression pattern is known [26]:
wg4~en1~hh1~ptc2,3,4~ci2,3,4~1 (on or expressed). The re-
maining nodes in every cell of the parasegment are set to 0 (off, or
not expressed). Overall, the dynamics of the SPN settles to one of
ten attractors – usually divided into four qualitatively distinct
groups, see Figure 4: (1) wild-type with three extra variations (PTC
mutant, double wg bands, double wg bands PTC mutant); (2)
Broad-stripe mutant; (3) No segmentation; and (4) Ectopic (with
the same variations as wild-type). Albert and Othmer estimated
that the number of configurations that converge to the wild-type
attractor is approximately 6|1011 – a very small portion of the
total number of possible configurations (&7|1016) – and that the
broad-stripe mutant attractor basin contains about 90% of all
possible configurations [26].
Figure 1. (A) LUT for Boolean automaton f ~x ^ (y _ z) and (B)
components of a single LUT entry.
doi:10.1371/journal.pone.0055946.g001
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
3
March 2013 | Volume 8 | Issue 3 | e55946
The inner and output nodes of each cell in a parasegment – that
is, every node except the input node SLP – that has reached a
stable configuration (attractor) are always in one of the following
five patterns.
N I1: all nodes are off except PTC, ci, CI and CIR.
N I2: same as I1 but states of ptc, PH, SMO, CIA and CIR are
negated.
N I3: all nodes are off except en, EN, hh, HH and SMO.
N I4: same as I3 but PTC and SMO are negated.
N I5: negation of I4, except PTC and CIR remain as in I4.
Figure 2. Connectivity graph of the SPN model. The fifteen genes and proteins considered in the SPN model are represented (white nodes).
The incoming edges to a node x originate in the nodes that are used by x to determine its transition. Shaded nodes represent the spatial signals
(states of WG, HH and hh in neighbouring cells). Note that SLP – derived from an upstream intra-cellular signal – is an input node to this network. The
self-connection it has represents the steady-state assumption: SLPtz1
i
~SLPt
i. Notice also that this graph represents the fully synchronous version of
the model, where modifications concerning PH and SMO have been made (see main text for details).
doi:10.1371/journal.pone.0055946.g002
Figure 3. A parasegment in the SPN model. Cells are represented horizontally, where the top (bottom) row is the most anterior (posterior) cell.
Each column is a gene, protein or complex – a node in the context of the BN model. The specific pattern shown corresponds to the wild-type initial
expression pattern observed at the onset of the segment polarity genes regulatory dynamics (xini); Black/on (white/off) squares represent expressed
(not expressed) genes or proteins.
doi:10.1371/journal.pone.0055946.g003
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
4
March 2013 | Volume 8 | Issue 3 | e55946
For example, the wild-type configuration corresponds – from
anterior to posterior cell – to the patterns I3, I2, I1 and I5. Thus
the pattern I4 is only seen in mutant expression patterns. The
patterns I1 to I5 can be seen as attractors of the inner- and
output-node dynamics of each cell in a parasegment.
Besides the fact that the SPN is probably the most well-known
discrete dynamical system model of biochemical regulation, we
chose it to exemplify our methodology because (1) it has been well-
validated experimentally, despite the assumption that genes and
proteins operate like on/off switches with synchronous transitions
and (2) the model includes both intra-cellular regulation and inter-
cellular signalling in a spatial array of cells. The intra and inter-
cellular interactions in the SPN model result in a dynamical
landscape that is too large to characterize via an STG, while
adding also an extra level of inter-cellular (spatial) regulation. The
ability to deal with such multi-level complexity makes our
methodology particularly useful. As we show below, we can
uncover the signals that control collective information processing
in such (spatial and non-spatial) complex dynamics.
Methodology and Results
Micro-level Canalization via Schemata
In previous work, we used schema redescription to demonstrate that
we can understand more about the dynamical behaviour of
automata networks by analysing the patterns of redundancy present
in their (automata) components (micro-level), rather than looking
solely at their macro-level or collective behaviour [39]. Here we
relate the redundancy removed via schema redescription with the
concept of canalization, and demonstrate that a characterization of
the full canalization present in biochemical networks leads to a
better understanding of how cells and collections of cells
‘compute’. Moreover, we show that this leads to a comprehensive
characterization of control in automata models of biochemical
regulation. Let us start by describing the schema redescription
methodology. Since a significant number of new concepts and
notations are introduced in this, and subsequent sections, a
succinct glossary of terms as well as a table with the mathematical
notations used is available in Data S1.
From the extended view of canalization introduced earlier, it
follows that the inputs of a given Boolean automaton do not
control its transitions equally. Indeed, substantial redundancy in
state-transition functions is expected. Therefore, filtering redun-
dancy out is equivalent to identifying the loci of control in
automata. In this section we focus on identifying the loci of control
in individual automata by characterizing the canalization present
in their transition functions. First, we consider how subsets of
inputs in specific state combinations make other inputs redundant.
Then we propose an additional form of canalization that accounts
for input permutations that leave a transition unchanged. Later, we
use this characterization of canalization and control in individual
automata to study networks of automata; this also allows us to
analyse robustness and collective computation in these networks.
Wildcard schemata and enputs.
Consider the example
automaton x in Figure 5A, where the entire subset of LUT entries
in F with transitions to on is depicted. This portion of entries in F
can be redescribed as a set of wildcard schemata, F’:ff ’ug. A wildcard
schema f ’u is exactly like a LUT entry, but allows an additional
wildcard symbol, # (also represented graphically in grey), to appear
in its condition (see Figure 5B). A wildcard input means that it
accepts any state, leaving the transition unchanged. In other words,
wildcard inputs are redundant given the non-wildcard input states
specified in a schema f ’u. More formally, when the truth value of
an input Boolean variable i in a schema f ’u is defined by the third
(wildcard) symbol, it is equivalent to stating that the truth value of
automaton x is unaffected by the truth value of i given the
conditions defined by f ’u: (xjf ’u,i)~(xjf ’u,:i). Each schema
Table 1. Boolean logic formulae representing the state-transition functions for each node in the SPN (four-cell parasegment)
model.
Index
Node
State{TransitionFunction
1
SLPtz1
i
/0 if i~1 _ i~2; 1 if i~3 _ i~4;
2
wgtz1
i
/(CIAt
i ^ SLPt
i ^ :CIRt
i) _ (wgt
i ^ (CIAt
i _ SLPt
i) ^ :CIRt
i)
3
WGtz1
i
/wgt
i
4
entz1
i
/(WGt
i{1 _ WGt
iz1) ^ :SLPt
i
5
ENtz1
i
/ent
i
6
hhtz1
i
/ENt
i ^ :CIRt
i
7
HHtz1
i
/hht
i
8
ptctz1
i
/CIAt
i ^ :ENt
i ^ :CIRt
i
9
PTCtz1
i
/ptct
i _ (PTCt
i ^ :HHt
i{1 ^ :HHt
i{1)
10
PHt
i
/PTCt
i ^ (HHt
i{1 _ HHt
iz1)
11
SMOt
i
/:PTCt
i _ (HHt
i{1 _ HHt
iz1)
12
citz1
i
/:ENt
i
13
CItz1
i
/cit
i
14
CIAtz1
i
/CIt
i ^ (:PTCt
i _ hht
i{1 _ hht
iz1 _ HHt
i{1 _ HHt
iz1)
15
CIRtz1
i
/CIt
i ^ PTCt
i ^ :hht
i{1 ^ :hht
iz1 ^ :HHt
i{1 ^ :HHt
iz1
The subscript represents the cell index; the superscript represents time. Note that every node has a numerical index assigned to it, which we use for easy referral
throughout the present work. The extra-cellular nodes, hh,HH and WG in adjacent cells are indexed as follows: 16 to 21 denote hhi{1, hhiz1, HHi{1, HHiz1, WGi{1
and WGiz1 in this order.
doi:10.1371/journal.pone.0055946.t001
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
5
March 2013 | Volume 8 | Issue 3 | e55946
redescribes a subset of entries in the original LUT, denoted by
Uu:ffa : fa
f 0
ug (
means ‘is redescribed by’).
Wildcard schemata are minimal in the sense that none of the
(non-wildcard) inputs in the condition of a schema can be ‘raised’
to the wildcard status and still ensure the automaton’s transition to
the
same
state.
Because
wildcard
schemata
are
minimal,
Uu6(Uw ^ Uw6(Uu, Vf ’u, f ’w[F’. In other words, a wildcard
schema is unique in the sense that the subset of LUT entries it
redescribes is not fully redescribed by any other schema. However,
in general Uu\Uw=1. This means that schemata can overlap in
terms
of
the
LUT
entries
they
describe.
In
Figure
5,
U1:ff1,f5,f9,f13g
and
U9:ff4,f5,f6,f7g,
therefore
U1\U9:ff5g. The set of wildcard schemata F’ is also complete.
This means that for a given LUT F there is one and only one set
F’ that contains all possible minimal and unique wildcard
schemata. Since wildcard schemata are minimal, unique and they
form a complete set F’, they are equivalent to the set of all prime
implicants obtained during the first step of the Quine & McCluskey
Boolean minimization algorithm [40]. Typically, prime implicants
are computed for the fraction of the LUT that specifies transitions
Figure 4. The ten attractors reached by the SPN. These attractors are divided in four groups: wild-type, broad-stripe, no segmentation and
ectopic. More specifically: (a) wild-type, (b) variant of (a), (c) wild-type with two wg stripes, (d) variant of (c), (e) broad-stripe, (f) no segmentation, (g)
ectopic, (h) variant of (g), (i) ectopic with two wg stripes, and (j) variant of (i). The wild-type attractor (a) is referred to as Awt in the results and
discussion sections.
doi:10.1371/journal.pone.0055946.g004
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
6
March 2013 | Volume 8 | Issue 3 | e55946
to on. Then a subset of the so-called essential prime implicants is
identified. The set of essential prime implicants is the subset of
prime implicants sufficient to describe (cover) every entry in the
input set of LUT entries. However, to study how to control the
transitions of automata we use the set of all prime implicants, since
it encodes every possible way a transition can take place. The set
F’ may also contain any original entry in F that could not be
subsumed by a wildcard schema. Although the upper bound on
the size of F’ is known to be O(3k=
ffiffiffi
k
p
) [41], the more input
redundancy there is, the smaller the cardinality of F’.
The condition of a wildcard schema can always be expressed as
a logical conjunction of literals (logical variables or their negation),
which correspond to its non-wildcard inputs. Since a wildcard
schema is a prime implicant, it follows that every literal is essential to
determine the automaton’s transition. Therefore, we refer to the
literals in a schema as its essential input states, or enputs for short. To
summarize, each enput in a schema is essential, and the
conjunction of its enputs is a sufficient condition to control the
automaton’s transition. It also follows that the set F’ of wildcard
schemata can be expressed as a disjunctive normal form (DNF) – that
is, a disjunction of conjunctions that specifies the list of sufficient
conditions to control automaton x, where each disjunction clause
is a schema. The DNF comprising all the prime implicants of a
Boolean function f is known as its Blake’s canonical form [42]. The
canonical form of f always preserves the input-output relationships
specified by its LUT F. Therefore, the basic laws of Boolean logic
– contradiction, excluded middle and de Morgan’s laws – are
preserved by the schema redescription.
Schema redescription is related to the work of John Holland on
condition/action rules to model inductive reasoning in cognitive
systems [43] and to the general RR framework proposed by Annette
Figure 5. Schema redescription. (A) Subset of LUT entries of an example automaton x that prescribe state transitions to on (1); white (black)
states are 0 (1). (B) Wildcard schema redescription; wildcards denoted also by grey states. Schema f ’9 is highlighted to identify the subset of LUT
entries U9:ff4,f5,f6,f7g it redescribes. (C) Two-symbol schema redescription, using the additional position-free symbol; the entire set F’ is
compressed into a single two-symbol schema: f ’’1. Any permutation of the inputs marked with the position-free symbol in f ’’1 results in a schema in
F’. Note that f ’’1 redescribes the entire set of entries with transition to on and thus jHhj~14. Since there is only one set of marked inputs, the
position-free symbol does not require an index. Although this figure depicts only the schemata obtained for the subset of LUT entries of x that
transition to on, entries that do not match any of these schemata transition to off (since x is a Boolean automaton).
doi:10.1371/journal.pone.0055946.g005
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
7
March 2013 | Volume 8 | Issue 3 | e55946
Karmiloff-Smith to explain the emergence of internal representa-
tions and external notations in human cognition [44]. Our
methodology to remove redundancy from automata LUTs also
bears similarities with the more general mask analysis developed by
George Klir in his ‘reconstructability’ analysis, which is applicable
to any type of variable [45]. In addition, prime implicants have
been known and used for the minimization of circuits in electrical
engineering since the notion was introduced by Quine &
McCluskey [40]; similar ideas were also used by Valiant [46]
when introducing Probably Approximately Correct (PAC) learning.
Two-symbol schemata.
We now introduce a different and
complementary form of redundancy in automata, which we
consider another form of canalization. Wildcard schemata identify
input states that are sufficient for controlling an automaton’s
transition (enputs). Now we identify subsets of inputs that can be
permuted in a schema without effect on the transition it defines
[39]. For this, a further redescription process takes as input the set
of wildcard schemata (F’) of x to compute a set of two-symbol
schemata F’’:ff ’’hg (see Figure 5C). The additional position-free
symbol (0
m) above inputs in the condition of a schema f ’’ means that
any subset of inputs thus marked can ‘switch places’ without affecting the
automaton’s transition. The index of the position-free symbol, when
necessary, is used to differentiate among distinct subsets of
‘permutable’ inputs. A two-symbol schema f ’’h redescribes a set
Hh:ffa : faf 00
h g of LUT entries of x; it also redescribes a subset
H’h(F’ of wildcard schemata.
A two-symbol schema f ’’h captures permutation redundancy in a set
of wildcard schemata H’h. More specifically, it identifies subsets of
input states whose permutations do not affect the truth value of the
condition, leaving the automaton’s transition unchanged. In group
theory, a permutation is defined as a bijective mapping of a non-
empty set onto itself; a permutation group is any set of
permutations of a set. Permutation groups that consist of all
possible permutations of a set are known as symmetric groups under
permutation [47]. For Boolean functions in general, the study of
permutation/symmetric groups dates back to Shannon [48] and
McCluskey [49] (see also [50]).
Two-symbol schemata identify subsets of wildcard schemata
that form symmetric groups. We refer to each such subset of input
states that can permute in a two-symbol schema – those marked
with the same position-free symbol – as a group-invariant enput. Note
that a group-invariant enput may include wildcard symbols
marked with a position-free symbol. More formally, a two-symbol
schema f ’’ can be expressed as a logical conjunction of enputs –
literal or group-invariant. Let us denote the set of literal enputs on
the condition of f ’’ by X‘(X – the non-wildcard inputs not
marked with the position-free symbol. For simplicity, n‘~ X‘
j
j.
A group-invariant enput g is defined by (1) a subset of input
variables Xg(X that are marked with an identical position-free
symbol, and (2) a permutation constraint (a bijective mapping) on Xg
defined by the expression ng~n0
gzn1
gzn#
g , where ng~ Xg
, n0
g is
the number of inputs in Xg in state 0 (off), and n1
g is the number of
inputs in Xg in state 1 (on). We further require that at least two of
the quantities n0
g,n1
g and n#
g are positive for any group-invariant
enput g. We can think of these two required positive quantities as
subconstraints; in particular, we define a group-invariant enput by
the two subconstraints n0
g,n1
g, since n#
g is always derivable from
those two given the expression for the overall permutation
constraint. This precludes the trivial case of subsets of inputs in
the same state from being considered a valid group-invariant
enput – even though they can permute leaving the transition
unchanged. A two-symbol schema f ’’ has n‘ literal enputs and g
group-invariant enputs; each of the latter type of enputs is defined
by a distinct permutation constraint for g~1,:::,g. An input
variable whose truth value is the wildcard symbol in a given
schema is never a literal enput (it is not essential by definition).
However, it can be part of a group-invariant enput, if it is marked
with a position-free symbol. Further details concerning the
computation of wildcard and two-symbol schemata are available
in Data S2.
In our working example, the resulting two-symbol schema (see
Figure 5C) contains n‘~2 literal inputs: X‘~fi2~0,i3~1g. It
also contains one (g~1) group-invariant enput Xg~fi1,i4,i5,i6g
with size ng~4 and subconstraints n0
g~1 ^ n1
g~1. This rede-
scription
reveals
that
the
automaton’s
transition
to
on
is
determined only by a subset of its six inputs: as long as inputs 2
and 3 are off and on, respectively, and among the others at least one is on and
another is off, the automaton will transition to on. These minimal control
constraints are not obvious in the original LUT and are visible
only after redescription.
We equate canalization with redundancy. The more redundancy
exists in the LUT of automaton x, as input-irrelevance or input-
symmetry (group-invariance), the more canalizing it is, and the
more compact its two-symbol redescription is, thus jF’’jvjFj. In
other words – after redescription – input and input-symmetry
redundancy in F is removed in the form of the two symbols. The
input states that remain are essential to determine the automaton’s
transition. Below we quantify these two types of redundancy,
leading to two new measures of canalization. Towards that, we
must first clearly separate the two forms of redundancy that exist
in 2-symbol schemata. The condition of a two-symbol schema f ’’
with a single group-invariant enput g – such as the one in
Figure 5C – can be expressed as:
^
ij[X0
‘
:ij
^
ij[X1
‘
ij ^
X
ij[Xg
:ij§n0
g
0
@
1
A ^
X
ij[Xg
ij§n1
g
0
@
1
A
ð1Þ
where X 0
‘ is the set of literal enputs that must be off, and X 1
‘ is the
set of literal enputs that must be on (thus X‘~X 1
‘ |X 0
‘ ). This
expression separates the contributions (as conjunctions) of the
literal enputs, and each subconstraint of a group-invariant enput.
Since we found no automaton in the target model (see below) with
schemata containing more than one group-invariant enput, for
simplicity and without lack of generality, we present here only this
case (g~1). See Data S3 for the general expression that accounts
for multiple group-invariant enputs (gw1).
All possible transitions of x to on are described by a set F1’’ of
two-symbol schemata. This set can also be expressed in a DNF,
where each disjunction clause is given by Expression 1 for all
schemata f ’’[F1’’: Transitions to off are defined by the negation of
such DNF expression: F0’’: :f ’’,Vf ’’[F1’’
f
g. Canalization of an
automaton x is now characterized in terms of two-symbol
schemata that capture two forms of redundancy: (1) input-irrelevance
and (2) input-symmetry (group-invariance). We next describe the
procedure to compute 2-symbol schemata for a an automaton x.
Readers not interested in the algorithmic details of this compu-
tation can safely move to the next subsection.
The procedure starts with the set of wildcard schemata F’
obtained via the first step of the Quine & McCluskey algorithm
[40] (see above). The set F’ is then partitioned into subsets H’i
such that,
F’~
[
i
H’i:
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
8
March 2013 | Volume 8 | Issue 3 | e55946
where each H’i contains every schema x’[F’ with equal number of
zeroes (n0), ones (n1), and wildcards (n#), with n0zn1zn#~k. In
other words, the H’i are equivalence classes induced on F’ by n0,
n1, and n#. This is a necessary condition for a set of wildcard
schemata to form a symmetric group. The algorithm then iterates
on each H’i, checking if it contains a symmetric group; i.e. if it
contains wildcard schemata with all the permutations of the largest
set of inputs variables possible. If it does, it marks those input
variables as a group-invariant enput in H’i and moves to another
subset H’j. If it does not, then it checks for symmetric groups in
smaller sets of input variables within each set H’i. It does this by
iteratively expanding the search space to include all subsets of H’i
with cardinality jH’ij{1. The procedure is repeated, if no
symmetric groups are found, until the subsets contain only one
wildcard schema.
Although several heuristics are implemented to prune the search
space, the algorithm is often not suitable for exhaustively searching
symmetric groups in large sets of schemata. However, the
individual automata found in models of biochemical regulation
and signalling networks typically have a fairly low number of
inputs. Therefore, schema redescription of their LUT leads to
manageable sets of wildcard schemata, which can be exhaustively
searched for symmetric groups. Indeed, as shown below, all
automata in the SPN model have been exhaustively redescribed
into two-symbol schemata. For additional details on the compu-
tation of schemata see Data S2.
Quantifying Canalization: Effective Connectivity and
Input Symmetry
Schemata uncover the ‘control logic’ of automata by making the
smallest input combinations that are necessary and sufficient to
determine transitions explicit. We equate canalization with the
redundancy present in this control logic: the smaller is the set of
inputs needed to control an automaton, the more redundancy
exists in its LUT and the more canalizing it is. This first type of
canalization is quantified by computing the mean number of
unnecessary inputs of automaton x, which we refer to as input
redundancy. An upper bound is given by,
kr(x)~
P
fa[F
max
h:fa[Hh
n#
h
jFj
ð2Þ
and a lower bound is given by:
kr(x)~
P
fa[F
min
h:fa[Hh
n#
h
jFj
ð3Þ
These expressions compute a mean number of irrelevant inputs
associated with the entries of the LUT F. The number of
irrelevant inputs in a schema f ’’h is the number of its wildcards n#
h .
Because each entry fa of F is redescribed by one or more schemata
f ’’h, there are various ways to compute a characteristic number of
irrelevant inputs associated with the entry, which is nonetheless
bounded by the maximum and minimum number of wildcards in
the set of schemata that redescribe fa. Therefore, the expressions
above identify all schemata f ’’h whose set of redescribed entries Hh
includes fa. The upper (lower) bound of input redundancy,
Equation 2 (Equation 3), corresponds to considering the maximum
(minimum) number of irrelevant inputs found for all schemata f ’’h
that redescribe entry fa of the LUT – an optimist (pessimist)
quantification of this type of canalization. Notice that input
redundancy is not an estimated value. Also, it weights equally each
entry of the LUT, which is the same as assuming that every
automaton input is equally likely.
Here we use solely the upper bound, which we refer to
henceforth simply as input redundancy with the notation kr(x). This
means that we assume that the most redundant schemata are
always accessible for control of the automaton. We will explore
elsewhere the range between the bounds, especially in regards to
predicting the dynamical behaviour of BNs. The range for input
redundancy is 0ƒkr(x)ƒk, where k is the number of inputs of x.
When kr(x)~k we have full input irrelevance, or maximum
canalization, which occurs only in the case of frozen-state
automata. If kr(x)~0, the state of every input is always needed
to determine the transition and we have no canalization in terms
of input redundancy.
In the context of a BN, if some inputs of a node x are irrelevant
from a control logic perspective, then its effective set of inputs is
smaller than its in-degree k. We can thus infer more about
effective control in a BN than what is apparent from looking at
structure alone (see analysis of macro-level control below). A very
intuitive way to quantify such effective control, is by computing the
mean number of inputs needed to determine the transitions of x,
which we refer to as its effective connectivity:
ke(x)~k(x){kr(x)
ð4Þ
whose range is 0ƒke(x)ƒk. In this case, ke(x)~0 means full
input irrelevance, or maximum canalization, and kr(x)~k, means
no canalization.
The type of canalization quantified by the input redundancy
and effective connectivity measures does not include the form of
permutation redundancy entailed by group-invariant enputs. Yet
this is a genuine form of redundancy involved in canalization, as in
the case of nested canalization [14], since it corresponds to the
case in which different inputs can be alternatively canalizing. The
two-symbol schema redescription allows us to measure this form of
redundancy by computing the mean number of inputs that
participate in group-invariant enputs, easily tallied by the
occurrence of the position-free symbol (0) in schemata. Thus we
define a measure of input symmetry for an automaton x, whose
upper-bound is given by
ks(x)~
P
fa[F
max
h:fa[Hh
n0
h
jFj
ð5Þ
and a lower-bound by,
ks(x)~
P
fa[F
min
h:fa[Hh
n0
h
jFj
ð6Þ
where n
0
h is the number of position-free symbols in schema f ’’h.
The upper bound of input symmetry (Equation 5) corresponds
to considering an optimist quantification of this type of canaliza-
tion. Here we use solely the upper bound, which we refer to
henceforth simply as input symmetry and denote by ks(x). Again,
the assumption is that the most redundant schemata are always
accessible for control of the automaton. The range for input
symmetry is 0ƒks(x)ƒk. High (low) values mean that permuta-
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
9
March 2013 | Volume 8 | Issue 3 | e55946
tions of input states are likely (unlikely) to leave the transition
unchanged.
Canalization in automata LUTs – the micro-level of networks of
automata – is then quantified by two types of redundancy: input
redundancy using kr(x) and input symmetry with ks(x). To be able to
compare the canalization in automata with distinct numbers of
inputs, we can compute relative measures of canalization:
k
r (x)~ kr(x)
k(x) ;
k
s (x)~ ks(x)
k(x)
ð7Þ
the range of which is ½0,1: Automata transition functions can have
different amounts of each form of canalization, which allows us to
consider four broad canalization classes for automata: class A with
high kr(x) and high ks(x), class B with high kr(x) and low ks(x),
class C with low kr(x) and high ks(x), and class D with low kr(x)
and low ks(x). We will explore these classes in more detail
elsewhere. Below, these measures are used to analyse micro-level
canalization in the SPN model and discuss the type of functions
encountered. Before that, let us introduce an alternative repre-
sentation of the canalized control logic of automata, which allows
us to compute network dynamics directly from the parsimonious
information provided by schemata.
Network Representation of a Schema
Canalization in an automaton, captured by a set of schemata,
can also be conveniently represented as a McCulloch & Pitts
threshold network – introduced in the 1940s to study computation
in interconnected simple logical units [51]. These networks consist
of binary units that can transition from quiescent to firing upon
reaching an activity threshold (t) of the firing of input units. To
use this type of network to represent two-symbol schemata we
resort to two types of units. One is the state unit (s-unit), which
represents an input variable in a specific Boolean state; the other is
the threshold unit (t-unit) that implements the condition that causes
the automaton to transition. Two s-units are always used to
represent
the
(Boolean)
states
of
any
input
variable
that
participates as enput in the condition of an automaton x: one
fires when the variable is on and the other when it is off. To avoid
contradiction, the two s-units for a given variable cannot fire
simultaneously. Directed fibres link (source) units to (end) units,
propagating a pulse – when the source unit is firing – that
contributes to the firing of the end unit. The simultaneous firing of
at least t (threshold) incoming s-units into a t-unit, causes the latter
to fire.
In the example automaton in Figure 5, the set of schemata F’’
contains only one schema. This schema can be directly converted
to a (2-layer) McCulloch & Pitts network. This conversion, which
is possible due to the separation of subconstraints given by
Expression (1), is shown in Figure 6 and explained in its caption.
Note that in the McCulloch & Pitts representation, the transition
of the automaton is determined in two steps. First, a layer of
threshold units is used to check that the literal and group-invariant
constraints are satisfied; then, a second layer – containing just one
threshold unit – fires when every subconstraint in Expression (1)
has been simultaneously satisfied, determining the transition. This
means that in this network representation each schema with literal
enputs and at least a group-invariant enput requires two layers and
three t-units. Since in McCulloch & Pitts networks each threshold
unit has a standard delay of one time step, this network
representation of a schema takes two time steps to compute its
transition.
We
introduce
an
alternative
threshold
network
representation of a two-symbol schema f ’’ that only requires a
single t-unit and takes a single time delay to compute a transition.
We refer to this variant as the Canalizing Map of a schema or CM
for short. A CM is essentially the same as a McCulloch and Pitts
network, with the following provisos concerning the ways in which
s-units and t-units can be connected:
1. Only one fibre originates from each s-unit that can participate
as enput in f ’’ and it must always end in the t-unit used to
encode f ’’.
2. The fibre that goes from a s-unit to the t-unit can branch out into
several fibre endings. This means that if the s-unit is firing, a
pulse propagates through its outgoing fibre and through its
branches. Branching fibres are used to capture group-invariant
enputs, as we explain later.
3. Branches from distinct s-units can fuse together into a single
fibre ending – the fused fibre increases the end t-unit’s firing
activity by one if at least one of the fused fibres has a pulse.
4. A fibre that originates in a t-unit encoding a schema f ’’ must
end in a s-unit that corresponds to the automaton transition
defined by f ’’.
Figure 7 depicts the elements of a single schema’s CM. Table 2
summarizes the rules that apply to the interconnections between
units. Transitions in CMs occur in the same way as in standard
McCulloch & Pitts networks. Once sufficient conditions for a
transition are observed at some time t, the transition occurs at
tz1. The firing (or not) of t-units is thus assumed to have a
standard delay (one time-step), identical for all t-units. Note that in
CMs, s-units can be regarded as a special type of t-unit with
threshold t~1 that send a pulse through their outgoing fibres
instantaneously. Next we describe the algorithm to obtain the CM
representation of a schema. Readers not interested in the
algorithmic details of this computation can safely bypass the next
subsection.
Algorithm
to
obtain
the
canalizing
map
of
a
schema.
Given a 2-symbol schema f ’’, there are two steps
involved in producing its CM representation. The first is
connecting s-units to the t-unit for f ’’ in such a way that it fires,
if and only if, the constraints of f ’’ – defined by Expression (1) –
are satisfied. The second step is determining the appropriate firing
threshold t for the t-unit. If the schema does not have group-
invariant enputs, the conversion is direct as for the standard
McCulloch &
Pitts network – see Figure 6: The
s-units
corresponding to literal enputs ij[X‘ are linked to the t-unit using
a single fibre from each s-unit to the t-unit, which has a threshold
t~n‘. If the schema has a group-invariant enput, its subcon-
straints are implemented by branching and fusing fibres connect-
ing the s-units and the t-unit. In cases such as our example
automaton
x
(Figures
5
and
6)
where the
subconstraints
n0
g~n1
g~1, the solution is still simple. To account for subcon-
straint n0
g, it is sufficient to take an outgoing fibre from each of the
s-units ij[Xg : ij~0 and fuse them into a single fibre ending.
Therefore, if at least one of these s-units is firing, the fused fibre
ending transmits a single pulse to the t-unit, signalling that the
subconstraint has been satisfied. Increasing the t-unit’s threshold
by one makes the t-unit respond to this signal appropriately. The
same applies for subconstraint n1
g, using a similar wiring for s-units
ij[Xg : ij~1. The final threshold for the t-unit that captures the
example schema of Figure 5 is thus t~n‘zn0
gzn1
g~2z1z1~4,
as shown in Figure 8C.
The case of general group-invariant constraints is more
intricate. Every literal enput ij[X‘ is linked to the t-unit via a
single fibre exactly as above. Afterwards, the subconstraints n0
g and
n1
g of a group-invariant enput g are treated separately and
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
10
March 2013 | Volume 8 | Issue 3 | e55946
consecutively. Note that for every input variable ij in the set Xg of
symmetric input variables, there are two s-units: one representing
ij in state 0 and another in state 1. To account for subconstraint n0
g
on the variables of set Xg, let S(Xg be the set of s-units that
represent the variables of the group-invariant enput that can be in
state 0, where jXgj~ng. Next, we identify all possible subsets
of S, whose cardinality is ng{(n0
g{1). That is, S~ Si : Si
f
x5S ^ jSij~ng{(n0
g{1)g. For each subset Si[S, we take an
outgoing fibre from every s-unit in it and fuse them into a single
fibre ending as input to the schema t-unit. After subconstraint n0
g is
integrated this way, the threshold of the t-unit is increased by,
jSj~
ng
ng{(n0
g{1)
!
~
ng
n0
g{1
!
ð8Þ
This procedure is repeated for the subconstraint n1
g on Xg. The
final threshold of the t-unit is,
t~n‘z
ng
n0
g{1
!
z
ng
n1
g{1
!
ð9Þ
This algorithm is illustrated for the integration of two example
subconstraints in Figure 9; in Figure 8, the case of the only schema
describing the transitions to on of running example automaton x is
shown. Further details concerning this procedure are provided in
Data S3.
The canalizing map of an automaton.
The algorithm to
convert a single schema f ’’ to a CM is subsequently used to
produce the CM of an entire Boolean automaton x as follows:
Each schema f ’’[F’’ is converted to its CM representation. Each
state of an input variable is represented by a single s-unit in the
resulting threshold network. In other words, there is a maximum
of two s-units (one for state 0 and one for state 1) for each input
variable that is either a literal enput or participates in a group-
invariant enput of x. The resulting threshold network is the
canalizing map of x. The connectivity rules of automata CMs
include the following provisos:
1. Every s-unit can be connected to a single t-unit with a single
outgoing fibre, which can be single or have branches.
2. Therefore, the number of outgoing fibres coming out of a s-unit
(before any branching) corresponds to the number of schemata
f ’’[F’’ in which the respective variable-state participates as an
enput. If such a variable is included in a group-invariant enput,
then the fibre may have branches.
3. Any subset set of t-units with threshold t~1 for the same
automaton transition (x~0 or x~1) are merged into a single t-
Figure 6. McCulloch & Pitts representation of Expression (1).
The conjunction clauses in Expression (1) for the example automaton x
are directly mapped onto a standard McCulloch & Pitts network with
two layers. On one layer the two literal enputs are accounted for by a
threshold unit (at the top) with threshold t~n‘~2. There is also a
group-invariant enput with permutation subconstraints on both
Boolean states. Two threshold units on the same layer are used to
capture these. The threshold unit on the left accounts for the
permutation subconstraint n1
g~1. It thus has as incoming s-units the
inputs xi[Xg : xi~1 and threshold t~n1
g~1. In a similar way, the
threshold unit on the right accounts for the subconstraint n0
g~1. When
all the constraints (literal and group-invariant) are satisfied, the last
threshold unit (second layer) ‘fires’ causing the transition to on.
doi:10.1371/journal.pone.0055946.g006
Figure 7. Elements of a Canalizing Map. Every s-unit is a circle,
labelled according the automaton’s input it represents and coloured
according to its state: black is on and white is off (here we use light-blue
for a generic state). The t-unit (schema) is represented using a larger
circle. One of its halves is coloured, and the other labelled with the t-
unit’s threshold t. Fibres can be single, or branched. In this example
there are branching fibres only, where fibre fusions represent all
possible combinations of two out of the three s-units.
doi:10.1371/journal.pone.0055946.g007
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
11
March 2013 | Volume 8 | Issue 3 | e55946
unit (also with t~1), which receives all incoming fibres of the
original t-units. In such scenario, any fused branches can also
be de-fused into single fibres. Note that this situation
corresponds to schemata that exhibit nested canalization,
where one of several inputs settles the transition, but which do
not form a symmetric group.
The CM of x can be constructed from the subset of schemata
F1’’ (the conditions to on), or F0’’ (the conditions to off). When the
conditions are not met for convergence to on, one is guaranteed
convergence to off (and vice-versa). However, since we are
interested in exploring scenarios with incomplete information
about the states of variables in networks of automata rather than a
single automaton (see below), we construct the CM of a Boolean
automaton x including all conditions, that is using F’’:F1’’|F0’’.
This facilitates the analysis of transition dynamics where automata
in a network can transition to either state. Figure 10 depicts the
complete CM of the example automaton x described in Figure 5–
now including also its transitions to off.
By uncovering the enputs of an automaton, we gain the ability
to compute its transition with incomplete information about the state of
every one of its inputs. For instance, the possible transitions of the
automaton in Figure 5 are fully described by the CM (and
schemata) in Figure 10; as shown, transitions can be determined
from a significantly small subset of the input variables in specific
state combinations. For instance, it is sufficient to observe i3~0 to
know that automaton x transitions to off. If x was used to model
the interactions that lead a gene to be expressed or not, it is easy to
see that to down-regulate its expression, it is sufficient to ensure
that the regulator i3 is not expressed. This is the essence of
canalization: the transition of an automaton is controlled by a
small subset of input states. In the macro-level canalization section
below, we use the CM’s ability to compute automata transitions
with incomplete information to construct an alternative portrait of
network dynamics, which we use in lieu of the original BN to study
collective dynamics. Let us first apply our micro-level methodology
to the SPN model.
Micro-level Canalization in the SPN Model
The automata in the SPN fall in two categories: those that have
a single input (k~1), the analysis of which is trivial, namely, SLP,
WG, EN, HH, ci and CI, and those with kw1. The two-symbol
schemata and canalization measures for each automaton in the
SPN model are depicted in Figure 11; Figure 12 maps the
automata to their canalization classes. Schemata easily display all
the sufficient combinations of input states (enputs) to control the
transitions of the automata in this model, which represent the
inhibition or expression of genes and proteins. Indeed, the
resulting list of schemata allows analysts to quickly infer how
control operates in each node of the network. Wildcard symbols
(depicted in Figure 11 as grey boxes) denote redundant inputs.
Position-free symbols (depicted in Figure 11 as circles), denote
‘functionally equivalent’ inputs; that is, sets of inputs that can be
alternatively used to ensure the same transition. For example, for
wg to be expressed, SLP, the previous state of wg (reinforcing
feedback loop) and CIA can be said to be ‘functionally equivalent’,
since any two of these three need to be expressed for wg to be
expressed. The several schemata that are listed for the expression
or inhibition of a specific node (genes and gene products), give
experts alternative ‘recipes’ available to control the node according
to the model – and which can be experimentally tested and
validated.
Let
us
now
present
some
relevant
observations
concerning micro-level canalization in the SPN model:
1. The inhibition of wg can be attained in one of two ways: either
two of the first three inputs (SLP, wg, CIA) are off
(unexpressed), or CIR is on (expressed). The expression of wg
– essential in the posterior cell of a parasegment to attain the
wild-type expression pattern (Figure 3)– is attained in just one
way: CIR must be off (unexpressed), and two of the other three
inputs (SLP, wg, CIA) must be on (expressed). Note the
simplicity of this control logic vis a vis the 24~16 possible
distinct ways to control wg specified by its LUT, given that it is
a function of 4 inputs. This control logic is also not obvious
from the Boolean logic expression of node wg, as shown in
Table 1; at the very least, the schemata obtained for wg
provide a more intuitive representation of control than the
logical expression. Moreover, schema redescription, unlike the
logical expression, allows us to directly quantify canalization.
The control logic of this gene shows fairly high degree of both
types of canalization: even though there are k~4 inputs, on
average, only ke~1:75 inputs are needed to control the
transition, and ks~2:25 inputs can permute without effect on
the transition (see Figures 11 and 12); wg is thus modelled by
an automaton of class A.
2. The inhibition of CIR can be attained in one of two simple,
highly canalized, ways: either one of its first two inputs (PTC,
CI) is off (unexpressed), or one of its four remaining inputs (hh
and HH in neighbouring cells) is on (expressed); all other
inputs can be in any other state. The expression of CIR can be
attained in only one specific, non-canalized, way: the first two
inputs must be on (expressed), and the remaining four inputs
must be off (unexpressed) – a similar expression behaviour is
found for hh and ptc. Note the simplicity of this control logic
vis a vis the 26~64 possible distinct ways to control CIR
specified by its LUT, given that it is a function of 6 inputs.
While, in this case, the control logic is also pretty clear from
the original Boolean logic expression of node CIR (in Table 1),
the schemata obtained for CIR provide a more intuitive
representation of control and allows us to directly quantify
canalization. CIR is a protein with a very high degree of both
types of canalization: even though there are k~6 inputs, on
average, only ke~1:08 inputs are needed to control the
transition, and ks~5:25 inputs can permute without effect on
Table 2. Connectivity rules in canalizing maps.
s-units
t-units
incoming fibres
one or more
one or more
outgoing fibres
one per schema of which is enput
one for the transition it causes
branching out
Yes
no
fusing in
No
yes
doi:10.1371/journal.pone.0055946.t002
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
12
March 2013 | Volume 8 | Issue 3 | e55946
the transition (see Figures 11 and 12). This high degree of
both types of canalization, which is not quantifiable directly
from the logical expression or the LUT, is notable in
Figure 12, where CIR emerges very clearly as an automaton
of class A.
3. The control logic of CIA entails high canalization of the input
redundancy kind. For instance, its inhibition can be achieved
by a single one of its six inputs (CI off) and its expression by two
inputs only (PTC off and CI on). On the other hand, there is low
canalization of the input symmetry kind, therefore CIA is
modelled by an automaton in class B.
4. The expression of en – essential in the anterior cell of a
parasegment to achieve the wild-type phenotype – depends on
the inhibition of (input node) SLP in the same cell, and on the
expression of the wingless protein in at least one neighbouring
cell.
Figure 8. Canalizing map of example automaton x character-
ized by a single schema. (A) Since f ’’ (shown on top) has n‘~2, the
corresponding s-units for literal enputs xi[X‘ are directly linked to the t-
unit for f ’’ with single fibres; t~n‘~2. (B) Adding the subconstraint
n0
g~1 of the group-invariant enput Xg~fi1,i4,i5,i6g. In this case,
ng{(n0
g{1)~ng~4, so there is only one subset Si(S and thus a single
branch from each s-unit in the group-invariant, fused into a single
ending. The threshold becomes t~n‘z
ng
n0
g{1
~2z
4
0
~3. (C)
Finally, we add the second subconstraint n1
g~1 of the group-invariant
enput Xg, which has the same properties as the subconstraint
integrated in (B). The final threshold of the t-unit is given by (9),
therefore t~n‘z
ng
n0
g{1
z
ng
n1
g{1
~2z
4
0
z
4
0
~4. Notice
that only the input-combinations that satisfy the constraints of
Expression (1) for f ’’ can lead to the firing of the t-unit; in other words,
the canalizing map is equivalent to schema f ’’.
doi:10.1371/journal.pone.0055946.g008
Figure 9. Procedure for obtaining the canalizing map of a
group-invariant subconstraint. (A) subconstraint defined by n0
g~2,
where ng~4. The first step is to consider the s-units (in state 0) for the
four input variables in the group invariant enput Xg~fi1,i2,i3,i4g. Next
we identify all the subsets Si of these s-units containing ng{(n0
g{1)~3
s-units: fi1,i2,i3g,fi1,i2,i4g,fi1,i3,i4g,fi2,i3,i4g (shown with dotted ar-
rows). From every s-unit in each such subset Si, we take an outgoing
fibre to be joined together into a single fibre ending as input to the t-
unit. Finally, we increase the threshold of the t-unit by the total number
of subsets, that is tA~
ng
n0
g{1
~
4
4
~4. (B) An example of the
same procedure but for n0
g~3 and ng~4: tB~
ng
n0
g{1
~
4
2
~6.
doi:10.1371/journal.pone.0055946.g009
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
13
March 2013 | Volume 8 | Issue 3 | e55946
5. Most automata in the model fall into canalization class B
described above. CIR and wg discussed above display greatest
input symmetry, and fall in class A (see Figure 12).
6. Looking at all the schemata obtained in Figure 11, we notice a
consistent pattern for all spatial signals, hhi+1, HHi+1 and
WGi+1. Whenever they are needed to control a transition
(when they are enputs in the schemata of other nodes), either
they are off in both neighbouring cells, or they are on in at least
one of the neighbouring cells. For instance, for a given cell i,
HH in neighbouring cells is only relevant if it is unexpressed in
both cells (HHi+1~0), or if it is expressed in at least one of
them (HHi{1~1 _ HHiz1~1). This means that the six nodes
corresponding to spatial signals affecting a cell in a paraseg-
ment can be consolidated into just three neighbour nodes, a similar
consolidation of spatial signals was used previously by
Willadsen & Wiles [52] to simplify the spatial model into a
single-cell non-spatial model. In what follows, we refer to these
spatial signals simply as nhh, nHH and nWG. If such a node is
off it means that the corresponding original nodes are off in both
adjacent cells; if it is on it means that at least one of the
corresponding original nodes in an adjacent cell is on.
7. Only PTC and wg have feedback loops that are active after
schema redescription, both for their inhibition and expression;
these are self-reinforcing, but also depend on other enputs (see
also Figures 13 and 14).
Because this is a relatively simple model, some of the
observations about control, especially for nodes with fewer inputs,
could be made simply by looking at the original transition
functions in Table 1, since they are available as very simple logical
expressions – this is the case of CIR, but certainly not wg above.
However, the quantification of canalization requires the additional
symbols used in schema redescription to identify redundancy,
which
are
not
available
in
the
original
automata
logical
expressions or their LUTs. Moreover, the transition functions of
automata in larger Boolean models of genetic regulation and
signalling are rarely available as simple logical expressions, and
nodes can be regulated by a large number of other nodes, thus
making such direct comprehension of control-logic difficult. In
contrast, since redescription uncovers canalization in the form of
input redundancy and symmetry, the more canalization exists, the
more redundancy is removed and the simpler will be the schemata
representation of the logic of an automaton. This makes canalizing
maps (CM) particularly useful, since they can be used to visualize
and compute the minimal control logic of automata. The CMs
that result from converting the schemata of each node in the SPN
to a threshold-network representation are shown in Figure 13 and
Figure 14. For a biochemical network of interest, such as the SPN
or much larger networks, domain experts (e.g. biomedical
scientists and systems and computational biologists) can easily
ascertain the control logic of each component of their model from
the schemata or the corresponding CMs.
In summary, there are several important benefits of schema
redescription of Boolean automata vis a vis the original Boolean
logic expression or the LUT of an automaton: (1) a parsimonious
and intuitive representation of the control logic of automata, since
redundancy is clearly identified in the form of the two additional
symbols, which gives us (2) the ability to quantify all forms of
canalization in the straightforward manner described above;
finally, as we elaborate next, the integration of the schema
redescription (or CMs) of individual automata in a network (micro-
level) allows us to (3) characterize macro-level dynamics parsimoniously,
uncovering minimal control patterns, robustness and the modules
responsible for collective computation in these networks.
Macro-level Canalization and Control in Automata
Networks
After removing redundancy from individual automata LUTs in
networks (micro-level), it becomes possible to integrate their
canalizing logic to understand control and collective dynamics of
automata networks (macro-level). In other words, it becomes
feasible to understand how biochemical networks process infor-
mation collectively – their emergent or collective computation
[39,53–56].
Dynamics
canalization
map
and
dynamical
modularity.
The CMs obtained for each automaton of a BN,
such as the SPN model (see Figures 13 and 14), can be integrated
into a single threshold network that represents the control logic of
the entire BN. This simple integration requires that (1) each
automaton is represented by two unique s-units, one for transition
to on and another to off, and (2) s-units are linked via t-units with
appropriate fibres, as specified by each individual CM. Therefore
a unique t-unit represents each schema obtained in the redescrip-
tion process. This results in the Dynamics Canalization Map (DCM)
for the entire BN. Since the DCM integrates the CMs of its
constituent automata, it can be used to identify the minimal control
conditions that are sufficient to produce transitions in the dynamics
of the entire network. Notice that when a node in the original BN
undergoes a state-transition, it means that at least one t-unit fires
in the DCM. When a t-unit fires, according to the control logic of
Figure 10. Canalizing Map of automaton x. (A) complete set of
schemata F’’ for x, including the transitions to on shown in Figure 5 and
the transitions of off (the negation of the first).(B) canalizing map; t-units
for schemata f ’’2 and f ’’3 were merged into a single t-unit with
threshold t~1 (see main text). (C) effective connectivity, input
symmetry and input redundancy of x.
doi:10.1371/journal.pone.0055946.g010
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
14
March 2013 | Volume 8 | Issue 3 | e55946
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
15
March 2013 | Volume 8 | Issue 3 | e55946
the DCM, it can cause subsequent firing of other t-units. This
allows the identification of the causal chains of transitions that are the
building blocks of macro-level dynamics and information processing,
as explained in detail below.
Another important feature of the DCM is its compact size.
While the dynamical landscape of an automata network, defined
by its state-transition graph (STG), grows exponentially with the
number of nodes – 2n in Boolean networks – its DCM grows only
linearly with 2n units plus the number of t-units needed (which is
the number of schemata obtained from redescribing every
automaton in the network): 2nz Pn
i~1 jF’’ij. Furthermore, the
computation of a DCM is tractable even for very large networks
with thousands of nodes, provided the in-degree of these nodes is
not very large. In our current implementation, we can exhaustively
perform schema redescription of automata with kƒkmax&20;
that is, LUTs containing up to 220 entries. It is very rare that
dynamical models of biochemical regulation have molecular
species that depend on more than twenty other variables (see
e.g. [57]). Therefore, this method can be used to study canalization
and control in all discrete models of biochemical regulation we
have encountered in the literature, which we will analyse
elsewhere.
It is important to emphasize that the integration of the CMs of
individual automata into the DCM does not change the control
logic encoded by each constituent CM, which is equivalent to the
logic encoded in the original LUT (after removal of redundancy).
Therefore, there is no danger of violating the logic encoded in the
original LUT of any automaton in a given BN. However, it is
necessary to ensure that any initial conditions specified in the
DCM do not violate the laws of contradiction and excluded
middle. This means, for instance, that no initial condition of the
DCM can have the two (on and off) s-units for the same automaton
firing simultaneously.
The DCM for a single cell in the SPN model is shown in
Figure 15. The spatial signals from adjacent cells are highlighted
using units with a double border (nhh,nHHandnWG). For the
simulations of the spatial SPN model described in subsequent
sections, we use four coupled single-cell DCMs (each as in
Figure 15) to represent the dynamics of the four-cell parasegment,
where nodes that enable inter-cellular regulatory interactions are
appropriately linked, as defined in the original model. Also, as in
the original model, we assume periodic boundary conditions for
the four-cell parasegment: the posterior cell is adjacent to the
anterior cell. When making inferences using the DCM, we use
signal to refer to the firing of a s-unit and the transmission of this
information through its output fibres. When a s-unit fires in the
DCM, it means that its corresponding automaton node in the
original BN transitioned to the state represented by the s-unit. We
also use pathway to refer to a logical sequence of signals in the
DCM.
We highlight two pathway modules in the DCM of the SPN in
Figure 15: M1 and M2. The first is a pathway initiated by either
the inhibition of WG in neighbour cells, or the expression of SLP
upstream in the same cell. That is, the initial pattern for this
module is M0
1~:nWG _ SLP. The initiating signal for M2 is
defined
by
the
negation
of
those
that
trigger
the
first:
M0
2~:M0
1~nWG ^ :SLP. Both modules follow from (external
or upstream) input signals to a single cell in the SPN; they do not
depend at all on the initial states of nodes (molecular species) of the
SPN inside a given cell. Yet, both of these very small set of initial
signals necessarily cause a cascade of other signals in the network
over time. M1 is the only pathway that leads to the inhibition of en
Figure 11. Micro-level canalization for the Automata in the SPN model. Schemata for inhibition (transitions to off) and expression
(transitions to on) are shown for each node (genes or proteins) in model. In-degree (k), input redundancy (kr), effective connectivity (ke), and input
symmetry (ks) are also shown.
doi:10.1371/journal.pone.0055946.g011
Figure 12. Quantification of canalization in the SPN automata. Relative input redundancy is measured in the horizontal axis (k
r ) and relative
input symmetry is measured in the vertical axis (k
r ). Most automata in the SPN fall in the class II quadrant, showing that most canalization is of the
input redundancy kind, though nodes such as CIR and wg display strong input symmetry too.
doi:10.1371/journal.pone.0055946.g012
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
16
March 2013 | Volume 8 | Issue 3 | e55946
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
17
March 2013 | Volume 8 | Issue 3 | e55946
(and EN) as well as the expression of ci (and CI). It also causes the
inhibition of hh and HH, both of which function as inter-cellular
signals for adjacent cells – this inhibition can be alternatively
controlled by the expression of CIR, which is not part of neither
M1 nor M2. Since M0
1 is a disjunction, its terms are equivalent:
either the inhibition of nWG or the upstream expression of SLP
control the same pathway, regardless of any other signals in the
network. M2 is the only pathway that leads to the expression of en
(and EN) as well as the inhibition of ci (and CI); It also causes the
inhibition of CIA, ptc and CIR – these inhibitions can be
alternatively controlled by other pathways. If the initial conditions
M0
2 are sustained for long enough (steady-state inputs), the
downstream inhibition of CIA and sustained inhibition of SLP
lead to the inhibition of wg (and WG); likewise, from sustaining
M0
2, the downstream expression of EN and inhibition of CIR lead
to the expression of hh (and HH). Since M0
2 is a conjunction, both
terms are required: both the expression of nWG and the upstream
inhibition of SLP are necessary and sufficient to control this
pathway module, regardless of any other signals in the network.
M1 and M2 capture a cascade of state transitions that are
inexorable once their initiating signals (M0
1 and M0
2) are observed:
M1~f:en,:EN, :hh, : HH, ci, CIg and M2~f:ci, :CI,
:CIA, :wg, :WG, :CIR, :ptc, en, EN, hh, HHg. Further-
more, these cascades are independent from the states of other nodes
in the network. As a consequence, the transitions within a module
are insensitive to delays once its initial conditions are set (and
maintained in the case of M2 as shown). The dynamics within these
portions of the DCM can thus be seen as modular; these pathway
modules can be decoupled from the remaining regulatory dynamics,
in the sense that they are not affected by the states of any other
nodes other than their initial conditions. Modularity in complex
networks has been typically defined as sub-graphs with high intra-
connectivity [21]. But such structural notion of community
structure does not capture the dynamically decoupled behaviour
of pathway modules such as M1 and M2 in the SPN. Indeed, it
has been recently emphasized that understanding modularity in
complex molecular networks requires accounting for dynamics
[58], and new measures of modularity in multivariate dynamical
systems have been proposed by our group [59]. We will describe
methods for automatic detection of dynamical modularity in
DCMs elsewhere.
Collective computation in the macro-level dynamics of autom-
ata networks ultimately relies on the interaction of these pathway
modules. Information gets integrated as modules interact with one
another, in such a way that the timing of module activity can have
an effect on downstream transitions. For instance, the expression
of CI via M1 can subsequently lead to the expression of CIA,
provided that nhh is expressed – and this is controlled by M2 in
the adjacent cells. The expression of CI can also be seen as a
necessary initial condition to the only pathway that results in the
expression of CIR, which also depends on the inhibition of nhh
and n HH and the expression of PTC, which in turn depends on
the interaction of other modules, and so on. As these examples
show, pathway modules allow us to uncover the building blocks of
macro-level control – the collective computation of automata
network models of biochemical regulation. We can use them, for
instance, to infer which components exert most control on a target
collective behaviour of interest, such as the wild-type expression
pattern in the SPN. Indeed, modules M1 and M2 in the SPN
model, which include a large proportion of nodes in the DCM,
highlight how much SLP and the spatial signals from neighbouring
cells control the dynamical behaviour of segment polarity gene
regulation in each individual cell. Particularly, they almost entirely
control the expression and inhibition of EN and WG; as discussed
further below. The behaviour of these proteins across a four-cell
parasegment mostly define the attractors of the model (including
wild-type). The transitions of intra-cellular nodes are thus more
controlled by the states of ‘external’ nodes than by the initial
pattern of expression of genes and proteins in the cell itself. This
emphasizes the well-known spatial constraints imposed on each
cell of the fruit fly’s developmental system [60,61]. We next study
and quantify this control in greater detail.
Dynamical unfolding.
A key advantage of the DCM is that
it allows us to study the behaviour of the underlying automata
network without the need to specify the state of all of its nodes.
Modules M1 and M2 are an example of how the control that a
very small subset of nodes exerts on the dynamics of SPN can be
studied. This can be done because, given the schema redescription
that defines the DCM, subsets of nodes can be assumed to be in an
unknown state. Since the schema redescription of every automaton
in the DCM is minimal and complete (see micro-level canalization
section), every possible transition that can occur is accounted for in
the DCM. By implementing the DCM as a threshold network, we
gain the ability to study the dynamics of the original BN by setting
the states of subsets of nodes. This allows us study convergence to
attractors, or other patterns of interest, from knowing just a few
nodes.
More formally, we refer to an initial pattern of interest of a BN
B as a partial configuration, and denote it by ^x. For example, M0
1 is a
partial configuration ^x~M0
1~SLP _ :nWG, where the states of
all other nodes is #, or unknown. We refer to dynamical unfolding as
the sequence of transitions that necessarily occur after an initial
partial configuration ^x, and denote it by s(^x)
P, where P is an
outcome pattern or configuration. From the DCM of the single-cell
SPN
model
(Figure
15),
we
have
s(M0
1)
M1
and
s(M0
2)
M2. An outcome pattern can be a fully specified
attractor A, but it can also be a partial configuration of an
attractor where some nodes remain unknown – for instance, to
study what determines the states of a specific subset of nodes of
interest in the network. In the first case, it can be said that ^x fully
controls the network dynamics towards attractor A. In the second,
control is exerted only on the subset of nodes with determined
logical states.
The ability to compute the dynamical unfolding of a BN from
partial configurations is a key benefit of the methodology
introduced here: it allows us to determine how much partial
configurations of interest control the collective dynamics of the
network. For instance, in the SPN model it is possible to
investigate how much the input nodes to the regulatory network
of each cell control its dynamics. Or, conversely, how much the
initial configuration of the intra-cellular regulatory network is
irrelevant to determining its attractor. The nodes within each cell
in a parasegment of the SPN are sensitive to three inter-cellular
(external) input signals: nWG, nhh and nHH, and one intra-
cellular (upstream) input, SLP. Given that the formation of
parasegment boundaries in D. melanogaster is known to be tightly
Figure 13. Canalizing Maps of individual nodes in the SPN model (part 1). The set of schemata for each automaton is converted into two
CMs: one representing the minimal control logic for its inhibition, and another for its expression. Note that nX denotes the state of node X in both
neighbour cells: :nXu:Xi{1 ^ :Xiz1 and nXuXi{1 _ Xiz1, where X is one of the spatial-signals hh, HH, or WG (see text).
doi:10.1371/journal.pone.0055946.g013
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
18
March 2013 | Volume 8 | Issue 3 | e55946
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
19
March 2013 | Volume 8 | Issue 3 | e55946
spatially constrained [60,61], it is relevant to investigate how
spatio-temporal control occurs in the SPN model. We already
studied the control power of SLP and nWG, which lead to
modules M1 and M2. We now exhaustively study the dynamical
unfolding of all possible states of the intra- and inter-cellular input
signals.
We assume that SLP (upstream) and the (external) spatial signals
are in steady-state to study what happens in a single cell. Since the
state of nHH is the same as nhh after one time step, we consolidate
those input signals into a single one: nhh. We are left with three
input signals to the intra-cellular regulatory network: nodes SLP,
nWG and nhh. Each of these three nodes can be in one of two
states (on, off) and thus there are eight possible combinations of
states for these nodes. Such simplification results in a non-spatial
model and this was done previously by Willadsen & Wiles [52].
Setting each such combination as the initial partial configuration
^x, and allowing the DCM to compute transitions, yields the results
shown in Figure 16. We can see that only two of the outcome
patterns reached by the eight input partial configurations are
ambiguous about which of the final five possible attractors is
reached. Each individual cell in a parasegment can only be in one
of five attractor patterns I1{I5 (see } background). This is the
case of groups G2 and G4 in Figure 16. For all the other input
partial configurations, the resulting outcome pattern determines
the final attractor. We also found that for almost every input
partial configuration, the states of most of the remaining nodes are
also resolved; in particular the nodes that define the signature of
the parasegment attractor – Engrailed (EN) and Wingless (WG) –
settle into a defined steady-state. Notice also that for two of the
input partial configurations (groups G3 and G5 in Figure 16), the
states of every node in the network settle into a fully defined
steady-state. The picture of dynamical unfolding from the intra-
and inter-cellular inputs of the single-cell SPN network also allows
us to see the roles played by modules M1 and M2 in the
dynamics. The six input configurations in groups G1, G2, and G3
depict the dynamics where M1 is involved, while the two input
configurations in G4 and G5 refer to M2 (node-states of each
module in these groups appear shaded in Figure 16). By
comparing the resulting dynamics, we can see clearly the effect
of the additional information provided by knowing if nhh is
expressed or inhibited; we also see that the dynamics of the
modules is unaffected by other nodes, as expected.
Figure 14. Canalizing Maps of individual nodes in the SPN model (cont). The set of schemata for each automaton is converted into two
CMs: one representing the minimal control logic for its inhibition, and another for its expression. Note that nX denotes the state of node X in both
neighbour cells: :nXu:Xi{1 ^ :Xiz1 and nXuXi{1 _ Xiz1, where X is one of the spatial-signals hh, HH, or WG (see text).
doi:10.1371/journal.pone.0055946.g014
Figure 15. Dynamics Canalization Map for a single cell of the SPN Model. Also depicted are pathway modules M1 (pink) and M2 (blue),
whose initial conditions depend exclusively on the expression and inhibition of input node SLP and of WG in neighbouring cells (the nWG spatial-
signals). M1~:nWG _ SLP, M2~:M1 (see details in text).
doi:10.1371/journal.pone.0055946.g015
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
20
March 2013 | Volume 8 | Issue 3 | e55946
It is clear from these results that (single-cell) cellular dynamics in
the SPN is almost entirely controlled from the inputs alone. We
can say that extensive micro-level canalization leads the macro-
level network dynamics to be highly canalized by external inputs –
a point we explore in more detail below. For the dynamical
unfolding depicted in Figure 16 we assumed that the three input
signals to the intra-cellular regulatory network are in steady-state,
focusing on a single cell. This is not entirely reasonable since inter-
cellular signals are regulated by spatio-temporal regulatory
dynamics in the full spatial SPN model. We thus now pursue
the identification of minimal partial configurations that guarantee
convergence to outcome patterns of interest in the spatial SPN
model, such as specific (parasegment) attractors.
Minimal configurations.
To automate the search of min-
imal configurations that converge to patterns of interest, we rely
again on the notion of schema redescription, but this time for
network-wide configurations rather than for individual automata
LUTs. Notice that the eight input partial configurations used in
the dynamical unfolding scenarios described in Figure 16 are
wildcard schemata of network configurations: the state of the 14
Figure 16. Dynamical unfolding of the (single-cell) SPN with partial input configurations. The eight initial partial configurations tested
correspond to the combinations of the steady-states of intra- and inter-cellular inputs SLP, nWG and nhh (and where nHH and nhh are merged into a
single node, nhh). The specific state-combinations of these three variables is depicted on the middle (white) tab of each dynamical unfolding plot.
Initial patterns that reach the same target pattern are grouped together in five groups G1 to G5 (identified in the top tab of each plot). The six input
configurations in groups G1, G2, and G3 depict the dynamics where pathway module M1 is involved (nodes involved in this module are highlighted
in pink.) The two input configurations in G4 and G5 depict the dynamics where pathway module M2 is involved (nodes involved in this module are
highlighted in blue.) Three of the eight combinations are in G1 because they reach the same final configuration which, although partial, can only
match the attractor I1. There are five possible attractor patterns of the SPN model for a single cell, shown in bottom right inset: I1 to I5 (see }
background). Attractors reached by each group are identified in the bottom tab of each plot. Groups G2 and G4 both unfold to an ambiguous target
pattern that can end in I2 or I5 for G2, and I3 or I4 for G4. Finally, the initial partial configurations in groups G3 and G5 are sufficient to resolve the
states of every node in the network.
doi:10.1371/journal.pone.0055946.g016
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
21
March 2013 | Volume 8 | Issue 3 | e55946
inner nodes is unknown (wildcard), and only three (input) nodes
(SLP, nWG,nhh) are set to a combination of Boolean states. Each
of these eight schemata redescribes 214 possible configurations of
the single-cell SPN. Six of the eight input schemata converge to
one of the five possible attractors for inner nodes in a single cell of
the SPN model (Figure 16). We can thus think of those six
schemata as minimal configurations (MCs) that guarantee conver-
gence to patterns (e.g. attractors) of interest.
More specifically, a MC is a 2-symbol schema x’’ that
redescribes a set of network configurations that converge to target
pattern P; when the MC is a wildcard schema, it is denoted by x’.
Therefore, s(x0’)
P. MC schemata, x’’ or x’, are network
configurations where the truth value of each constituent autom-
aton can be 0, 1, or # (unknown); symmetry groups are allowed
for x’’ and identified with position-free symbols 0m (see Micro-level
canalization section). An MC schema redescribes a subset H of the
set of configurations X: H:fx[X : x
x00g. A partial config-
uration is a MC if no Boolean state in it can be raised to the
unknown state (#) and still guarantee that the resulting partial
configuration converges to P. In the case of a two-symbol schema,
no group-invariant enput can be enlarged (include additional
node-states) and still guarantee convergence to P. Finally, the
target pattern P can be a specific network configuration (e.g. an
attractor), or it can be a set of configurations of interest (e.g. when
only some genes or proteins are expressed). After redescription of a
set of configurations X of a BN – a subset or its full dynamical
landscape – we obtain a set of two-symbol MCs X’’; a set of
wildcard MCs is denoted by X’. Similarly to micro-level schemata,
we can speak of enputs of MCs. In this context, they refer to
individual and sets of node-states in the network that are essential
to guarantee convergence to a target pattern.
The dynamical unfolding example of the single-cell SPN model
shows that to converge to the attractor I1 (Figure 16, G1), only the
states of the three input nodes need to be specified, in one of three
possible Boolean combinations: 000,100 or 110 for the nodes SLP,
nWG and nhh; all other (inner) nodes may be unknown (#).
Moreover, these three initial patterns can be further redescribed
into two schemata: X’~ff#,0,0g,f1,#,0gg. This shows that to
guarantee converge to I1, we only need to know the state of two
(input) nodes: either nWG ~nhh~0, or SLP = 1 and nhh~0. All
other nodes in the single-cell model can remain unknown.
Therefore, the MCs for attractor pattern I1 are:
X0~f###############00,##############1#0g ð10Þ
where the order of the inner nodes is the same as in Figure 16,
and the last three nodes are SLP, nWG and nhh in that order.
Notice that in this case there is no group-invariance, so X’’~X’.
Any initial configuration not redescribed by X’, does not converge
to pattern I1. Therefore, these MCs reveal the enputs (minimal set
of node-states) that control network dynamics towards attractor I1:
nhh must remain unexpressed, and we must have either SLP
expressed, or nWG unexpressed. However, as mentioned above,
this example refers to the case when the three input nodes are in
steady-state. For the single-cell SPN, the steady-state assumption is
reasonable. But for the spatial SPN, with parasegments of four
cells, we cannot be certain that the spatial signals (nWG and nhh)
have reached a steady-state at the start of the dynamics. Therefore,
we now introduce a procedure for obtaining MCs, without the
steady-state assumption, which we apply to the spatial SPN
network model.
It was discussed previously that individual automata in BN
models of biochemical regulation and signalling very rarely have
large numbers of input variables. This allows tractable computa-
tion of two-symbol schema redescription of their LUTs (see micro-
level section). In contrast, computing MCs for network configu-
rations easily becomes more computationally challenging. Even for
fairly small networks with n&20, the size of their dynamical
landscape becomes too large to allow full enumeration of the
possible configurations and the transitions between them. As
shown above, it is possible to identify pathway modules, and to
compute dynamical unfolding on the DCM, without knowing the
STG of very large BNs, but it remains not feasible to fully
redescribe their entire dynamical landscape.
One way to deal with high-dimensional spaces is to resort to
stochastic search (see e.g. [62]). We use stochastic search to obtain
MCs that are guaranteed to converge to a pattern of interest P.
We start with a seed configuration known to converge to P. Next, a
random node in a Boolean state is picked, and changed to the
unknown state. The resulting partial configuration is then allowed
to unfold to determine if it still converges to P. If it does, the
modified configuration becomes the new seed. The process is
repeated until no more nodes can be ‘raised’ to the unknown state
and still ensure convergence to P. Otherwise, the search continues
picking other nodes. The output of this algorithm (detailed in Data
S4) is thus a single wildcard MC. Afterwards, the goal is to search
for sets of MCs that converge to P. We do this in two steps: first we
search for a set of MCs derived from a single seed, followed by a
search of the space of possible different seeds that still converge to
P. We use two ‘tolerance’ parameters to determine when to stop
searching. The first, d, specifies the number of times a single seed
must be ‘reused’ in the first step. When the algorithm has reused
the seed d consecutive times without finding any new MCs, the
first step of the MC search stops. The second tolerance parameter,
r, is used to specify when to stop searching for new seeds from
which to derive MCs. When r consecutively generated random
(and different) seeds are found to be already redescribed by the
current set of MCs, the algorithm stops. Both parameters are reset
to zero every time a new MC is identified. These two steps are
explained in greater detail in Data S4.
The two-step stochastic search process results in a set of
wildcard schemata X’ that redescribe a given set of configurations
X guaranteed to converge to pattern P. We next obtain a set of
two-symbol MCs X’’ from X’, by identifying group-invariant
subsets of nodes using the same method described in the micro-
level canalization section. Since X’ can be quite large (see below),
this computation can become challenging. In this case, we restrict
the search for symmetric groups in X’ that redescribe a minimum
number b of wildcard MCs x’.
Notice that it is the DCM, implemented as a threshold network,
that allows us to pursue this stochastic search of MCs. With the
original BN, we cannot study dynamics without setting every
automaton to a specific Boolean truth value. With the DCM,
obtained from micro-level canalization, we are able to set nodes to
the unknown state and study the dynamical unfolding of a partial
configuration (see previous subsection) to establish convergence to
a pattern of interest. Therefore, the DCM helps us link micro-level
canalization to macro-level behaviour. Let us exemplify the
approach with the SPN model.
We started our study of MCs in the spatial SPN model, with a
set of seed configurations Xbio that contains the known initial
configuration of the SPN (shown in Figure 3), the wild-type
attractor (Figure 4a), and the five configurations in the dynamic
trajectory between them. When searching for MCs using these
seed configurations we set d~105. This resulted in a set containing
90 wildcard MCs X’bio (available in data S7). Using the set X’bio,
we performed the two-step stochastic search with r~106 and
(10)
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
22
March 2013 | Volume 8 | Issue 3 | e55946
d~105. This resulted in a much larger set of 1745 wildcard MCs
(available in data S8) which guarantee convergence to wild-type:
X’wt6X’bio. The number of literal enputs in each MC contained
in this set varies from 23 to 33 – out of the total 60 nodes in a
parasegment. In other words, from all configurations in X’wt we
can ascertain that to guarantee convergence to the wild-type
attractor, we need only to control the state of a minimum of 23
and a maximum of 33 of the 60 nodes in the network.
Equivalently, 27 to 37 nodes are irrelevant in steering the
dynamics of the model to the wild-type attractor – a high degree of
canalization we quantify below.
We chose to study two further subsets of X’wt separately: X’noP
and X’min. The first (available in data S9) is the subset of MCs that
do not have enputs representing expressed (on) proteins, except
SLP3,4 – since SLP in cells 3 and 4 is assumed to be present from
the start, as determined by the pair-rule gene family (see [26] and
introductory section). This is a subset of interest because it
corresponds to the expected control of the SPN at the start of the
segment-polarity dynamics, including its known initial configura-
tion (Figure 3); thus X’noP5X’wt. The second, X’min5X’wt is the
subset of MCs with the smallest number of enputs (available in data
S10. This corresponds to the set of 32 MCs in X’wt that have only
23 enputs each. This is a subset of interest because it allows us to
study how the unfolding to wild-type can be guaranteed with the
smallest possible number of enputs. Notice that X’min redescribes a
large subset of configurations in Xwt because it contains the MCs
with most redundant number of nodes. These sets of wildcard
MCs are available in data S7,S8, S9 and S10; Table 3 contains their
size.
There are severe computational limitations to counting exactly
the number of configurations redescribed by each set of MCs,
since it depends on using the inclusion/exclusion principle [63] to
count the elements of intersecting sets (MCs redescribe overlap-
ping sets of configurations). See Data S6 for further details. We can
report the exact value for jX’noPj~8:35|1010, which is about
14% of the number of configurations – or pre-patterns – estimated
by Albert & Othmer [26] to converge to the wild-type attractor
(6|1011). Using the inclusion/exclusion principle, it was also
computationally feasible to count the configurations redescribed
by a sample of 20 of the 32 MCs in X’min : 9:6|1011. Since this
sample of 20 MCs is a subset of X’min, which is a subset of X’wt,
we thus demonstrate that jXwtj§jXminj§9:6|1011, which is 1:6
times larger than the previously estimated number of pre-patterns
converging to the wild-type attractor [26]. This means that the
wild-type attraction basin is considerably (at least 1.6 times) larger
than previously estimated, with a lower bound of at least
9:6|1011 network configurations. Although it was not computa-
tionally feasible to provide exact counts for the remaining MC sets,
it is reasonable to conclude that the set X’wt redescribes a
significant proportion of the wild-type attractor basin, given the
number of configurations redescribed by 20 of its most canalized
MCs in comparison to the previous estimate of its size. Indeed, we
pursued a very wide stochastic search with large tolerance
parameters, arriving at a large number (1745) MCs, each of
which redescribes a very large set of configurations. For instance,
each MC with the smallest number of enputs (23) alone redescribes
1:37|1011 configurations, which is about 23% of the original
estimated size of the wild-type attractor basin, and 14% of the
lower bound for the size of the attractor basin we computed above.
Given the large number of MCs in the X’wt set, even with likely
large overlaps of configurations, much of the attractor basin ought
to be redescribed by this set.
From X’wt, we derived two-symbol MC sets using b~8. That is,
due to the computational limitations discussed previously, we
restricted the search to only those two-symbol MCs x’’ that
redescribe at least b~8 wildcard MCs x’. Given that configura-
tions of the spatial SPN are defined by 60 automata states, the
group-invariance enputs we may have missed with this constraint
are rather trivial. For instance, we may have missed MCs with a
single group-invariant enput of 3 variables (any group-invariant
enput with 4 variables would be found), or MCs with 2 distinct
group-invariant enputs of 2 variables each (any MCs with 3 group-
invariant enputs would be found.) With this constraint on the
search for two-symbol MCs, we identified only the pair of two-
symbol MCs depicted in Figure 17: fx’’1,x’’2g – each redescribing
16 wildcard MCs – the MCs redescribed are available in data S13.
These two MCs redescribe 1:95|1011 configurations; that is,
about 32% of the wild-type attraction basin as estimated by [26],
or 20% of the lower bound for the size of the attractor basin we
computed above – a very substantial subset of the wild-type
attractor basin.
No other two-symbol MCs redescribing at least eight wildcard
MCs were found in the set X’wt. Therefore, X’’wt is comprised of
the wildcard MCs in X’wt with the addition of fx’’1,x’’2g and
removal of the wildcard MCs these two schemata redescribe.
Table 3 contains the size of all MC sets. Moreover, fx’’1,x’’2g
have no intersecting schemata with the additional three subsets of
X’’wt we studied. This means that the two-symbol redescription
(with b~8) is equal to the wildcard redescription of the sets of
configurations Xbio, XnoP and Xmin. The pair of two-symbol MCs
identified denote two very similar minimal patterns that guarantee
convergence to the wild-type attractor. In both MCs, the pairs of
nodes wg2,4, HH2,4 as well as ci4 and CI4 are marked with distinct
position-free symbols. In other words, they have three identical
group-invariant enputs. For x’’1 a fourth group-invariant enput
comprises the nodes hh1,3, while for x’’2 the fourth group-invariant
enput contains the nodes HH1,3. For x’’2 there is an extra literal
enput: ptc4~0 (ptc gene in fourth cell is unexpressed). The
Table 3. Macro-level canalization in the wildcard MC sets converging to wild-type in the SPN.
MC set
jX’’j
e (min)
e (max)
ne
nr
ns
X’wt
1745
23
33
24:01+0:08
35.99 +0:17
0:98+0:03
X’min
32
23
23
23+0
37 +0
0
X’bio
90
25
28
25:75+0:11
34.25 +0:11
0
X’noP
24
26
30
26:2+0:04
34.8 +0:04
0
The table lists for every set of MCs reported in the main text: cardinality, minimum number of enputs, maximum number of enputs, estimated canalization. Canalization
measures were obtained, for each MC set, from 10 independent samples of 104 configurations, thus j^Xj~105. Values shown refer to the mean plus 95% confidence
intervals for the 10 independent measurements.
doi:10.1371/journal.pone.0055946.t003
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
23
March 2013 | Volume 8 | Issue 3 | e55946
remaining literal enputs are identical to those of x’’1. The group-
invariance in these MCs is not very surprising considering the
equivalent roles of neighbouring hedgehog and Wingless for intra-
cellular dynamics – as discussed previously when the SPN’s DCM
was analysed. Notice that most group-invariance occurs for the
same genes or proteins in alternative cells of the parasegment; for
instance, wg expressed in either cell 2 or cell 4. Nonetheless, both
two-symbol MCs offer two minimal conditions to guarantee
convergence to the wild-type attractor, which includes a very large
proportion of the wild-type attractor basin. Therefore, they serve
as a parsimonious prescription for analysts who wish to control the
macro-level behaviour (i.e. attractor behaviour) of this system.
Finally,
the
MCs
obtained
observe
substantial
macro-level
canalization which we quantify below.
Quantifying Macro-level Canalization
In the micro-level canalization section, we defined measures of
input redundancy, effective connectivity and input symmetry to quantify
micro-level canalization from the schema redescription of individ-
ual automata. Since we can also redescribe configurations that
produce network dynamics, leading to the minimal configurations
(MCs) of the previous section, we can use very similar measures to
quantify macro-level canalization and control. At the macro-level,
high canalization means that network dynamics are more easily
controllable: MCs contain fewer necessary and sufficient node-
states (enputs) to guarantee convergence to an attractor or target
pattern P. Similarly to the micro-level case, we first define upper
and lower bounds of node redundancy computed from the set of MCs
X’’ for a target pattern:
nr(X,P)~
P
x[X
max
h:x[Hh
n#
h
jXj
ð11Þ
nr(X,P)~
P
x[X
min
h:x[Hh
n#
h
jXj
ð12Þ
These expressions tally the mean number of irrelevant nodes in
controlling network dynamics towards P for all configurations x of
a set of configurations of interest X (e.g. a basin of attraction). The
number of irrelevant nodes in a given MC x’’h is the number of its
wildcards n#
h . Because each configuration x is redescribed by one
or more MCs, there are various ways to compute a characteristic
number of irrelevant nodes associated with the configurations,
which is nonetheless bounded by the maximum and minimum
number of wildcards in the set of MCs that redescribe x.
Therefore, the expressions above identify all MCs whose set of
redescribed configurations Hh includes x. The upper (lower)
bound
of
node
redundancy,
Equation
11
(Equation
12),
corresponds to considering the maximum (minimum) number of
irrelevant nodes found for all MCs that redescribe configuration x
of the interest set – an optimist (pessimist) quantification of this
type of macro-level canalization. Here we use solely the upper
bound, which we refer to henceforth simply as node redundancy with
the notation nr(X,P). Similarly to the micro-level case, the
assumption is that the most redundant MCs are always accessible
for control of the network towards pattern P. The range for node
redundancy is 0ƒnrƒn, where n is the number of nodes in the
network. When nr(X,P)~n we have full node irrelevance, or
maximum canalization, which occurs only in the case of networks
where the state of every node is not dependent on any input (that
is, when kr~k for every node). If nr(X,P)~0, the state of every
node is always needed to determine convergence to P and we have
no macro-level canalization.
Figure 17. Two-Symbol schemata with largest number of position-free symbols, obtained from redescription of Xwt. The pair
fx’’1,x’’1g were the two-symbol schemata obtained in our stochastic search; both include 4 pairs of symmetric node-pairs, each denoted by a circle
and a numerical index.
doi:10.1371/journal.pone.0055946.g017
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
24
March 2013 | Volume 8 | Issue 3 | e55946
If some nodes of a network are irrelevant to steer dynamics to P,
from a control logic perspective, we can say that P is effectively
controlled by a subset of nodes of the network with fewer than n
nodes. In other words, by integrating the micro-level control logic
of automata in a network into the DCM, we are able to compute
MCs and infer from those the macro-level effective control, which is
not apparent from looking at connectivity structure alone:
ne(X,P)~n{nr(X,P)
ð13Þ
whose range is 0ƒneƒn. If ne(X,P)~0 it means full node
irrelevance, or maximum canalization. When ne(X,P)~n, it
means no canalization i.e. one needs to control all n nodes to
guarantee converge to P.
Macro-level canalization can also manifest alternative control
mechanisms. The two-symbol schema redescription allows us to
measure this form of control by computing the mean number of
nodes that participate in group-invariant enputs, easily tallied by
the number of position-free symbols (n
0
h) in MC schemata x’’h that
characterize convergence to target pattern P. Thus, we quantify
the upper and lower bounds of node symmetry in a set of
configurations of interest X related to target pattern P (e.g. a
basin of attraction).
ns(X,P)~
P
x[X
max
h:x[Hh
n0
h
jXj
ð14Þ
ns(X,P)~
P
x[X
min
h:x[Hh
n0
h
jXj
ð15Þ
Here we use solely the upper bound, which we refer to
henceforth simply as node symmetry and denote by ns(X,P); its
range is ½0,n. Again, the assumption is that the most canalized
MCs are always accessible for control of the network towards
pattern P. High (low) values mean that permutations of node-
states are likely (unlikely) to leave the transition unchanged.
Macro-level canalization in network dynamics is then quantified
by two types of redundancy: node redundancy (or its counterpart,
effective control) and node symmetry. To be able to compare
macro-level control in automata networks of different sizes, we can
compute relative measures of canalization:
n
r(X,P)~ nr(X,P)
n
;
n
e(X,P)~ ne(X,P)
n
;
n
s(X,P)~ ns(X,P)
n
ð16Þ
whose range is ½0,1: Network dynamics towards a pattern of
interest P can have different amounts of each form of canalization,
which allows us to consider four broad classes of control in
network dynamics – just like the micro-level canalization case (see
above).
The two MCs identified above for the single-cell SPN model
(Eq. 10), redescribe the full set of configurations that converge to
I1. Since these MC schemata do not have group-invariant enputs,
node symmetry does not exist: ns(X,I1)~0. Node redundancy
and effective control is nr(X,I1)~15 and ne(X,I1)~2, respec-
tively. In other words, even though the network of the single-cell
SPN model comprises n~17 nodes, to control its dynamics
towards attractor I1, it is sufficient to ensure that the states of only
two nodes remain fixed; the initial state of the other 15 nodes is
irrelevant. More concretely, nhh must remain off and either SLP
remains on or nwg remains off. The relative measures become:
n
r(X,I1)~15=17 (&88% of nodes are redundant to guarantee
convergence to attractor I1) n
e(X,I1)~2=17 (one only needs to
control &12% of nodes to guarantee convergence to attractor I1),
and n
s(X,I1)~0 (there is no node symmetry in these MCs). This
means that there is a large amount of macro-level canalization of
the node redundancy type – and thus higher controllability – in
the basins of attraction of the SPN model where pattern I1 is
present.
The macro-level canalization measures above assume that the
interest set of configurations X can be enumerated. Moreover,
schema redescription of network configurations itself assumes that
X can be sufficiently sampled with our stochastic search method
(see previous sub-section). The node symmetry measure addition-
ally assumes that the set of wildcard MCs obtained by stochastic
search is not too large to compute symmetric groups. While these
assumptions are easily met for micro-level analysis, because LUT
entries of individual automata in models of biochemical regulation
do not have very large number of inputs, they are more
challenging at the macro-level. Certainly, canalization in the
single-cell SPN model can be fully studied at both the micro- and
macro-levels – see Figures 11 and 12 for the former as well as
example above for the latter. But quantification of macro-level
canalization of larger networks, such as the spatial SPN model,
needs to be estimated. Therefore, in formulae 11, 12, 14, and 15,
the set of configurations X is sampled: ^X. Configurations for ^X
are sampled from each MC in the set X’’, proportionally to the
number of configurations redescribed by each MC – i.e. roulette
wheel sampling. Configurations from a selected MC are sampled
by ascribing Boolean truth values to every wildcard in the MC
schema; the proportion of each of the truth values is sampled from
a uniform distribution. If a selected MC is a 2-symbol schema, the
truth-values of group-invariant enputs are also sampled from a
uniform distribution of all possible possibilities. Naturally, the
same configuration x can be redescribed by more than one MC h.
In summary, macro-level canalization for larger networks is
quantified with the estimated measures: ^nr, ^ne, and ^ns, as well as
their relative versions.
Tables 3 and 4 summarize the quantification of macro-level
canalization estimated for the four MC sets obtained above: X’’wt,
X’’min, X’’bio, and X’’noP. Effective control (ne) ranges between 23
and 26:2 nodes (out of 60) for the four sets of MCs; this means (see
n
e) that only 38 to 44% of nodes need to be controlled to
guarantee convergence to wild-type. This shows that there is
substantial macro-level canalization in the wild-type attractor
basin; from n
r, we can see that 56 to 62% of nodes are, on average,
redundant to guarantee convergence to wild-type. On the other
hand, macro-level canalization in the form of alternative (or
symmetric) control mechanisms is not very relevant on this
attractor basin, as observed by the low values of ns and n
s: in the
wild-type attractor basin, on average, only approximately 1 out 60
nodes, or 1:6% can permute.
Enput Power and Critical Nodes
Every MC is a schema, and hence comprises a unique set of
enputs, not entirely redescribed by any other MC. As defined in
the micro-level canalization section, an enput e can be literal – a
single node in a specific Boolean state – or a group-invariant
enput: a set of nodes with a symmetry constraint. Every enput e in
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
25
March 2013 | Volume 8 | Issue 3 | e55946
a given MC is essential to ensure convergence to a pattern P, e.g.
an attractor A. Consequently, if the state or constraint of e is
disrupted in the MC, without gaining additional knowledge about
the configuration of the network, we cannot guarantee conver-
gence to P. How critical is e in a set of configurations X redescribed
by an MC set X’’ – such as the set of MCs that redescribe a basin
of attraction? Since there are usually alternative MCs that
redescribe the possible dynamic trajectories to P, the more e
appears in X’’, the more critical it is in guaranteeing convergence
to P.
For instance, in the two MCs shown in Equation 10, the enput
e:(nhh~0) is common to both. Therefore, disrupting it, without
gaining additional knowledge about the state of other nodes,
would no longer guarantee convergence to the attractor pattern I1
in the single-cell SPN dynamics. Similarly, for the two-symbol MC
set of the spatial SPN model, shown in Figure 17, enputs
e:(hh2,4~0) and group-invariant enput e:(wg2~1 _ wg4~1)
appear in both MCs. Disrupting them, would no longer guarantee
convergence to wild-type attractor in the spatial SPN dynamics.
Let us quantify the potential disruption of target dynamics by
perturbation of enputs in an MC set. The power of an enput e in a
set of configurations X
X00 : s(x)
P, Vx[X, is given by:
(e,X00,P)~ jXej
jXj
ð17Þ
where Xe(X is the subset of configurations redescribed by X’’
that contain enput e: Xe:fx[X : xx00 ^ e[x00g. Thus, this
measure yields the proportion of configurations in X redescribed
by the MCs in which e is an enput; its range is ½0,1. If an enput
appears in every MC, as in the examples above, then E~1 – in
which case e is said to have full power over X’’. For the analysis of
the SPN model below when 0:5ƒEv1, e is a high power enput,
when 0vev0:5 it is a low power enput, and when E~0 it is a null
power enput. The larger the power of e, the more its perturbation is
likely to disrupt convergence to the target pattern P. When X is
too large, we estimate ^ – similarly to the canalization measures
discussed in the previous subsection.
We studied the wild-type attractor basin of the spatial SPN
model using the four MC sets of interest: X’’wt, X’’min, X’’bio, and
X’’noP (see Minimal configurations subsection above) focusing on
the power of literal enputs only. It is also possible to compute the
enput power of group-invariant enputs. For example, the two-
symbol MC x’’1 in Figure 17, has one of its four group-invariant
enputs defined by ci~1 _ CI~1. The power of this enput would
tally those MCs in which this condition holds. Nonetheless, here
we only measure the power of literal enputs and present the study
of the power of group-invariant enputs elsewhere. The enput
power computed for these four sets is depicted in Figure 18, where
the output nodes PH and SMO are omitted because they are
never input variables to any node in the SPN model, and therefore
have null power. For the discussion of these results, it is useful to
compare them to the known initial condition, xini depicted in
Figure 3, and the wild-type attractor, Awt depicted in Figure 4 (a).
Enput power in X’’wt (see Figure 18A). The enputs with full
power (E~1) are: SLP1,2~0, SLP3,4~1, hh2,4~0 and ptc1~0.
This is not entirely surprising since all of these genes and proteins
are specified as such in both xini and Awt. However, these values
show that these enputs must remain in these states in the entire
(sampled) wild-type basin of attraction. In other words, these
enputs are critical controllers of the dynamics to the wild-type
attractor. Indeed, the wild-type is not robust to changes in these
enputs, which are likely to steer the dynamics to other attractors,
as discussed further in the next section. Therefore, the spatial SPN
model appears to be unable to recover the dynamic trajectory to
the wild-type attractor when either the hedgehog gene is expressed
in cells two and four; or the patched gene is expressed in the
anterior cell, as well when the initial expression pattern of SLP
determined upstream by the pair-rule gene family is disrupted in
any way. There are also enputs with high power to control wild-type
behaviour:
wg1,3~WG1,3~0,
en1~1,
PTC1~0,
en2,4~0,
ptc3~1, CI3~0 and CIR3~1. Again, these are the states of
these genes and proteins in the known initial configuration of the
SPN xini, and most of them, except for ptc3~1, CI3~0 and
CIR3~1 correspond to their final states in Awt.
In Figure 18A every node in the SPN – except the omitted
nodes PH and SMO – appear as an enput, in at least one Boolean
state, in many cases with very low values of . Thus, while macro-
level dynamics is significantly canalized (see above), especially by
SLP and the spatial signals for each cell, control of wild-type can
derive from alternative strategies, whereby every node can act as
an enput in some context. Nonetheless, most nodes ultimately do
not observe much power to control wild-type behaviour, thus
interventions to disturb wild-type behaviour are most effective via
the few more powerful controllers (see also next section).
We can also compare the enput power computed for X’’wt
(Figure 18A), with the two-symbol MCs x’’1 and x’’2 in Figure 17.
These two MCs redescribe a significant portion of the wild-type
attractor basin – 20% of our lower bound count of this basin.
Because they only appear in X’’wt and not in any of the other MC
sets we studied, the portion of the wild-type attractor basin they
redescribe is unique to Xwt, and can be analysed via x’’1 and x’’2.
Most of the literal enputs specified in x’’1 and x’’2 have high power
in X’’wt, except for WG2~wg4~CIR1,2,4~1, which are enputs in
these two-symbol MCs that have low power. Conversely, there are
literal enputs with high-power in X’’wt that are not enputs in these
two-symbol MCs: EN2,4~0 and PTC1~0. A key distinguishing
feature of x’’1 and x’’2 is the expression of CIR across the entire
parasegment as well as of the wingless protein in the second cell,
both of which are different from the trajectory between the known
initial condition of the SPN and the wild-type attractor. Therefore,
x’’1 and x’’2 redescribe a (large) portion of the attractor basin
outside of the more commonly studied dynamical trajectories.
Enput power in X’’min (see Figure 18B). We found an
unexpected expression of CIR2~1 (now with full power) as well as
wg2~WG2~1 (high power). Other enputs whose expression is in
opposition to both xini and Awt appear with low power: HH2,4~1
and CIR1~1. This again suggests that there is a substantial subset
of the wild-type attractor basin, controlled by these and other
Table 4. Macro-level canalization in the wildcard MC sets
converging to wild-type in the SPN.
MC set
ne
nr
ns
X’wt
0.4 +0:001
0.6 +0:001
0.016 +0:002
X’min
0.38
0.62
0
X’bio
0.43 +0:001
0.57 +0:001
0
X’noP
0.436 +0:0007
0.564 +0:0007
0
The table lists the relative canalization measures for every set of MCs reported
in the main text. Canalization measures were obtained, for each MC set, from 10
independent samples of 104 configurations, thus j^Xj~105. Values shown refer
to the mean plus 95% confidence intervals for the 10 independent
measurements.
doi:10.1371/journal.pone.0055946.t004
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
26
March 2013 | Volume 8 | Issue 3 | e55946
enputs, distinct from the trajectory that results from the known
(biologically plausible) initial configuration. We can also see that
there is a significant number of nodes that do not play the role of
enput in any MC – nodes with null power, depicted as small grey
circles – as well as many more enputs with full power. X’’min
redescribes wild-type dynamics with the smallest number (23) of
enputs; this set contains only 32 MCs out of the 1731 in X’’wt.
However,
these
are
the
most
macro-canalizing
MCs
that
guarantee convergence to wild-type. Indeed, because of their
parsimony, they redescribe a very large subset of the wild-type
attractor basin with at least 1.6 times more configurations than
what was previously estimated for this basin (see above).
Therefore, X’’min provides a solid baseline for the understanding
of control in the wild-type attractor basin. This means that the
genes and proteins with full power in this set are critical controllers
of wild-type behaviour.
Enput power in X’’bio (see Figure 18C). Because this MC set
only redescribes configurations in the dynamic trajectory from xini
to Awt, the transient dynamics observed in X’’wt and X’’min, e.g.
wg2~1 and CIR2~1, disappear. There are, however, other
enputs with full power: wg1,3~WG1,3~0, en2,4~EN2,4~0,
ptc1~PTC1~0. These critical enputs are particularly important
Figure 18. Enput power in the wild-type basin of attraction of the spatial SPN model. Enput power is shown for each of the four sets of
MCs considered in our analysis: (A) X’’wt, (B) X’’min, (C) X’’bio and (D) X’’noP. A parasegment is represented by four rounded rectangles, one for each
cell, where the anterior cell is at the top, and posterior at the bottom. Since enput power is computed for every node in each of its two possible
states, every cell rectangle has two rows of circles. The bottom row (marked on the sides with a white circle on the outside) corresponds to enput
power of the nodes when off, while the top row is the enput power when the same nodes are on (marked on the sides with a dark circle). Each circle
inside a cell’s rectangle corresponds to the power of a given enput in the corresponding subset of MCs identified by the letters A to D. Full power is
highlighted in red, other values in blue and scaled, while null power is depicted using small grey circles. Full power occurs only for enputs that are
present in every MC (and configurations) of the respective set, whereas null power identifies nodes that are never enputs in any MC – always
irrelevant for the respective dynamical behaviour.
doi:10.1371/journal.pone.0055946.g018
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
27
March 2013 | Volume 8 | Issue 3 | e55946
for restricting analysis to a better-known portion of the wild-type
attractor basin, for which the model was especially built.
Enput power in X’’noP (see Figure 18D). This set of MCs is
useful to understand the beginning of the segment polarity
regulatory dynamics, with no proteins expressed. The set of
critical genes that must be expressed (on) are ptc3 and wg4, which
appear
with full power; moreover, en1~hh1~ptc2~ci2~1
appear with high power. As shown in the figure, most other
enputs with full or high power correspond to genes and proteins
that must be inhibited (off), except, of course, SLP3,4 that are
assumed to be always on in the SPN model.
We compared these results with previous work on identifying
critical nodes in the SPN model. Chaves et al. [38] deduced, from
the model’s logic, minimal ‘pre-patterns’ for the initial configura-
tion of the SPN that guarantee convergence to wild-type attractor.
More specifically, two necessary conditions and one sufficient
condition were deduced, which we now contrast with the enput
power analysis.
The first necessary condition for convergence to the wild-
type
attractor
is:
ptc3~1,
assuming
that
all
proteins
are
unexpressed (off ) initially, and the sloppy pair gene rule is
maintained constant (i.e. SLP1,2~0 ^ SLP3,4~1.) Of the MC sets
we analysed, only X’’noP
obeys the (biologically plausible)
assumptions for this necessary condition. As we can see in
Figure 18D, the enput ptc3~1 has full power on this MC set,
which confirms this previous theoretical result. However, since
every enput with full power is a necessary condition for the set of
configurations described by its MC set, we can derive other
necessary conditions for this set of configurations (with the same
assumptions), such as ptc1~0, wg3~0, or wg4~1 (see below). We
can also see that not all assumptions for the first necessary
condition are necessary; while the sloppy pair rule appears as four
enputs with full power, not all proteins are required to be
unexpressed: the expression of HH is irrelevant in every cell of the
parasegment, as is the expression of PTC2,3, WG2,4, CIA4, and
CIR1,2,3. Moreover, the enput power analysis allows us to identify
‘degrees of necessity’; some enputs may not be necessary, but
almost always necessary. This is the case of the expression of en1,
which has high power in X’’noP, but is not a necessary condition as
a few MCs can guarantee convergence to wild-type with en1~0
(which also appears as enput with low power). Naturally, if we
relax the assumptions for condition ptc3~1, it may no longer be a
necessary condition. This can be see when we look at the enput
power analysis of the entire (sampled) wild-type basin X’’wt
(Figure 18A) or the smaller X’’bio (Figure 18C). In these cases,
which still preserve the sloppy pair rule assumption, ptc3~1 is no
longer an enput with full power. This means that, according to this
model, if some proteins are expressed initially, ptc3~1 is no longer
a necessary condition. Interestingly, we found that in the most
macro-canalizing subset of the attractor basin, X’’min (Figure 18B)
– which assumes the sloppy pair rule constraint but is not
constrained to initially unexpressed proteins – ptc3~1 does
appear as an enput with full power again. This means that in the
most parsimonious means to control convergence to wild-type
attractor, ptc3~1 is a necessary condition too. It is noteworthy
that in this case, not only can some proteins be expressed, but the
expression of CIR2 is also a necessary condition (enput with full
power).
The second necessary condition for convergence to the
wild-type attractor is: wg4~1 _ en1~1 _ ci4~1, assuming that all
proteins are unexpressed (off) initially, and the sloppy pair gene
rule is maintained constant (i.e. SLP1,2~0 ^ SLP3,4~1) [38].
Again, only X’’noP obeys the (biologically likely) assumptions for
this necessary condition. As we can see in Figure 18D, the enput
wg4~1 has full power, therefore it is a necessary condition.
However, the enput en1~1 has high power, and the enput ci4~1
has no power. This means that they are not necessary, though
en1~1 is most often needed. These results suggest that this
necessary condition could be shortened to wg4~1, because in our
sampling of the wild-type attractor basin, in the subset meeting the
assumptions of the condition, we did not find a single configura-
tion where wg4~0. Even though our stochastic search was very
large, it is possible that there may be configurations, with no
proteins
expressed,
where
wg4~0 ^ (en1~1 _ ci4~1),
thus
maintaining the original necessary condition. However, our enput
power analysis gives a more realistic and nuanced picture of
control in the SPN model under the same assumptions. While the
necessary
condition
may
be
wg4~1 _ en1~1 _ ci4~1,
the
individual enputs have strikingly different power in controlling
for wild-type behaviour: ci4~1 was never needed (no power),
en1~1 has high power, and wg4~1 has full power. Naturally, if
we relax the assumptions for this condition, it may no longer be a
necessary condition. For instance, if we allow proteins to be
expressed initially (still preserving the sloppy pair constraint), we
can
find
MCs
that
redescribe
configurations
where
wg4~en1~ci4~0. We found 171 MCs in X’’wt (available in data
S14 where this condition is not necessary, one of them depicted in
Figure 19.
The sufficient condition for convergence to the wild-type
attractor is: wg4~1 ^ ptc3~1, assuming that the sloppy pair
gene rule is maintained constant (i.e. SLP1,2~0 ^ SLP3,4~1). A
variation of this sufficient condition assumes instead (maintaining
the sloppy pair gene rule): wg4~1 ^ PTC3~1 In their analysis,
Chaves et al. [38] assume that all proteins are unexpressed and
that many other genes are initially inhibited (off ). Even though in
Chaves et al. [38] the initial condition itself only requires
ptc1~ci1,3~0, the argument hinges on propositions and facts
that require knowing the state of additional genes such as
en2~wg3~hh2,4~0. While Chaves et al. [38] concluded rightly
from this minimal pre-pattern, that convergence to the wild-type
pattern has a remarkable error correcting ability to expression
delays in all other genes, the condition does not really describe
robustness to premature expression of genes and proteins. It is
interesting to investigate sufficient conditions that do require the
states of most variables to be specified, giving us the ability to study
robustness to both delays and premature expression of chemical
species. The MC schemata we obtained with our macro-level
analysis allows us to investigate such sufficient conditions directly.
We searched the entire MC set X’’wt to retrieve the MCs with
the fewest number of enputs specified as on. The 10 MCs (available
in S11) we retrieved contain only 26 literal enputs, where in six
MCs the two nodes in the sufficient condition above (wg4,ptc3),
plus the nodes from the sloppy pair rule (SLP3,4) are on, 24 are off
and the remaining 32 are wildcards, and thus irrelevant. In the
remaining MCs, instead of ptc3~1, we found PTC3~1 to be an
enput. In those MCs ptc3~#. Converting all wildcards to off in
one of these MCs, confirms the sufficient condition, as can be seen
from Figure 20A, where SLP3,4~wg4~ptc3~1, and everything
else is off. This can be seen as an ‘extreme’ condition to wild-type
attractor, with a minimum set of genes expressed. We also
searched for the opposite extreme scenario, retrieving all MCs
with the largest number of on nodes, that still converges to the
wild-type pattern (available in data S12. By replacing all wildcards
in such MCs to on, we obtained the configuration in which only 16
nodes must be inhibited (off ), while the remaining 44 are
expressed (on), depicted in Figure 20B. Interestingly, in this
extreme configuration, hh must remain off across the whole
parasegment.
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
28
March 2013 | Volume 8 | Issue 3 | e55946
Robustness to Enput Disruption
The power measure introduced in the previous subsection
allows us to predict critical nodes in controlling network dynamics
to a pattern of interest P. A natural next step is to investigate what
happens when the critical controllers are actually disrupted. We
can disrupt an enput e in an MC set with a variety of dynamic
regimes. Here, we adopt the approach proposed by Helikar et al.
[64], where a node of interest flips its state at time t with a
probability f, which can be seen to represent noise in regulatory
and signalling events, as well as the ‘concentration’ of a gene (its
corresponding mRNA) or protein – thus making it possible to use
Boolean networks to study continuous changes in concentration of
biochemical systems (see [64]).
We start from an initial set of configurations of interest: X 0.
This can be a single configuration, such as the known initial
configuration of the SPN X 0:fxinig (as in Figure 3A), where the
enput e is in a specific (Boolean) value. Next, we set the value of
noise parameter f, which is the probability that e momentarily flips
from its state in X 0 at time t. This noise is applied at every time
step of the simulated dynamics; when a state-flip occurs at time t,
the node returns to its original state at tz1 when noise with
probability f occurs again. Noise is applied to e from t~0 to t~m.
At time step t~mz1 no more noise is applied to e (f~0) and the
network is allowed to converge to an attractor. This process is
repeated for M trials. Finally, we record the proportions of the M
trials that converged to different attractors.
Since in this paper we only computed enput power for literal
enputs (see previous subsection), we also only study literal enput
disruption. It is straightforward to disrupt group-invariant enputs;
for instance, the group-invariant enput defined by ci~1_ CI ~1
from the two-symbol MC x’’1 in Figure 17, can be perturbed by
making ci~0^ CI ~0. Nonetheless, for simplicity, we present the
study of the disruption of group-invariant enputs elsewhere.
The enput power analysis in the previous subsection, revealed
that in the wild-type attractor basin (Xwt) of the spatial SPN model
there are the following critical nodes (or key controllers): across the
parasegment, SLP proteins must be inhibited in cells 1 and 2
(SLP1,2~0) and expressed in cells 3 and 4 (SLP3,4~1), as
determined by the pair-rule gene family; hedgehog genes (spatial
signals) in cells 2 and 4 must be inhibited (hh2,4~0); the patched
gene in the anterior cell must also be inhibited (ptc1~0). With the
stochastic intervention procedure just described, we seek to answer two
questions about these key controllers: (1) how sensitive are they to
varying degrees of stochastic noise? and (2) which and how many
other attractors become reachable when they are disrupted? In
addition to the seven full power enputs, for comparison purposes,
we also test the low power enput CI4~0. In the original SPN
model the states of SLP1,2,3,4
are fixed (the sloppy gene
constraints). Because these naturally become enputs with full
power (see Figure 18), it is relevant to include them in this study of
enput disruption. However, by relaxing the fixed-state constraint
on
SLP1,2,3,4, by inducing
stochastic
noise, the
dynamical
landscape of the spatial SPN model is enlarged from 256 to 260
configurations. This means that more attractors than the ten
identified for the SPN Boolean model (depicted in Figure 4) are
possible, and indeed found as explained below.
We used X 0:fxinig as the initial state of the networks analysed
via stochastic interventions, because of its biological relevance.
The simulations where performed with the following parameters:
f[½0:05,0:95, swept with D(f)~0:05, plus extremum values
f~0:02 and f~0:98; m~500 steps; M~104. The simulation
results are shown in Figure 21.
The first striking result is that disruption of SLP1~0 makes it
possible to drive the dynamics away from wild-type into one of five
other attractors (one of which a variant of wild-type). For fw0:15
no further convergence to wild-type is observed, and at f~0:05
the proportion of trials that converged to wild-type was already
very small. We also found phase transitions associated with the
values of f. For fƒ0:15 most trials converged to wild-type, wild-
type (ptc mutant), broad-stripes or no-segmentation, and a very
small proportion to two variants of the ectopic mutant. When
f~0:15 the proportion of trials converging to broad-stripes
reaches its peak, and decreases, so that no trial converged to this
mutant expression pattern for f§0:55. Finally, for f§0:55
convergence to the ectopic variants reaches its peak and decreases
steadily but does not disappear, while convergence to the no-
segmentation mutant increases becoming almost 100% when
f~0:98. We thus conclude that SLP1~0 is a wild-type attractor
enput which is very sensitive to noise.
In the case of SLP3~1, we observed convergence to an
attractor that is not any of the original ten attractors –
characterized by having two engrailed bands in cells 1 and 3
(see Data S5). The proportion of trials converging to wild-type and
to the new attractor decrease and increase respectively, reaching
similar proportions when f~0:5. When f~0:98, almost every
trial converged to the new attractor. We conclude that SLP3~1 is
a wild-type attractor enput whose robustness is proportional to
noise.
Disruption of SLP4~1 resulted in a behaviour similar to SLP1,
but with fewer possible attractors reached. As f is increased, fewer
trials converge to wild-type and growing proportions of trials
converge to the wild-type ptc mutant pattern (reaching a peak at
f~0:5) and the no-segmentation mutant. For more extreme values
of f, the majority of trials converged to the no-segmentation
mutant. However, an important difference with respect to SLP1
was observed: for fƒ0:5 the majority of trials converged to wild-
type, and convergence to this attractor is observed for the whole
range of f. Thus the wild-type phenotype in the SPN model is
much more robust to perturbations to the expression of SLP in the
posterior cell (SLP4~1), than to perturbations to its inhibition in
the anterior cell (SLP1~0).
With the parameters chosen, the disruption of SLP2~0 leads to
a remarkable similar behaviour: any disruption (any amount of
noise) leads to the same wild-type variant attractor pattern with
two wingless stripes (c). Therefore, SLP2~0 is not robust at all –
though the resulting attractor is always the same and a variant of
wild-type. In this case, convergence to a single attractor for all
values of f is the result of setting m~500 in our experiments.
When we lower the value of m enough in our simulations, for low
values of f, there are trials that are not perturbed and thus
maintain
convergence
to
the
wild-type
attractor.
But
any
Figure 19. A MC not requiring wg4~1 _ en1~1 _ ci4~1 in wild-
type attractor basin. When proteins are allowed to be expressed
initially, the second necessary condition, reported in [38], ceases to be a
necessary condition, as discussed in the main text; in the MC shown,
wg4, en1 and ci4 can be in any state and the network still converges to
the wild-type attractor.
doi:10.1371/journal.pone.0055946.g019
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
29
March 2013 | Volume 8 | Issue 3 | e55946
perturbation of SLP2~0 that occurs leads the dynamics to the
wild-type variant.
Disruption of hh2,4~0 increasingly drives dynamics to the
broad-stripes mutant. However, disruption of hh2 reveals greater
robustness since a large number of trials still converges to wild-type
for fƒ0:15, and residual convergence to wild-type is observed up
to f~0:75. In contrast, any disruption of hh4 above f~0:05 leads
to the broad-stripes mutant, and even very small amounts of
disruption lead to a large proportion of mutants. Similarly,
disruption of e:ptc1~0 drives the dynamics to one – and the
same – of the wild-type variants. Yet, when f~0:02 there is a
minute proportion of trajectories that still converge to the wild-
type attractor. Therefore, as expected, the wild-type attractor in
the SPN model is not very robust to disruptions of the enputs with
full power. Finally, and in contrast, no disruption of low-power
enput CI4~0 is capable of altering convergence to the wild-type
attractor.
Discussion
We introduced wildcard and two-symbol redescription as a
means to characterize the control logic of the automata used to
Figure 20. ‘Extreme’ configurations converging to wild-type in the SPN model. (A) A configuration with the minimal number of nodes
expressed that converges to wild-type, and its corresponding MC: 32 nodes are irrelevant, 24 must be unexpressed (off), and only 4 must be
expressed (on). (B) The opposite extreme condition where 16 genes and proteins are unexpressed and all other 44 are expressed.
doi:10.1371/journal.pone.0055946.g020
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
30
March 2013 | Volume 8 | Issue 3 | e55946
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
31
March 2013 | Volume 8 | Issue 3 | e55946
model networks of biochemical regulation and signalling. We do
this by generalizing the concept of canalization, which becomes
synonymous with redundancy in the logic of automata. The two-
symbol schemata we propose capture two forms of logical
redundancy, and therefore of canalization: input redundancy
and symmetry. This allowed us to provide a straightforward way
to quantify canalization of individual automata (micro-level), and to
integrate the entire canalizing logic of an automata network into
the Dynamics Canalization Map (DCM). A great merit of the
DCM is that it allows us to make inferences about collective
(macro-level) dynamics of networks from the micro-level canaliz-
ing logic of individual automata – with incomplete information.
This is important because even medium-sized automata models of
biochemical regulation lead to dynamical landscapes that are too
large to compute. In contrast, the DCM scales linearly with
number of automata – and schema redescription, based on
computation of prime implicants – is easy to compute for
individual automata with the number of inputs typically used in
the literature.
With this methodology, we are thus providing a method to link
micro- to macro-level dynamics – a crux of complexity. Indeed, in
this paper we showed how to uncover dynamical modularity: separable
building blocks of macro-level dynamics. This an entirely distinct
concept from community structure in networks, and allows us to
study complex networks with node dynamics – rather than just their
connectivity structure. The identification of such modules in the
dynamics of networks is entirely novel and provides insight as to
how the collective dynamics of biochemical networks uses these
building blocks to produce its phenotypic behaviour – towards the
goal of explaining how biochemical networks ‘compute’.
By basing our methodology on the redescription of individual
automata (micro-level), we also avoid the scaling problems faced
by previous schemata approaches which focused solely on
redescription of the dynamical landscape (macro-level) of networks
[52]. By implementing the DCM as a threshold network, we show
that we can compute the dynamical behaviour of the original
automata network from information about the state of just a few
network nodes (partial information). In its original formulation, the
dynamic unfolding of an automata network cannot be computed
unless an initial state of all its nodes is specified. In turn, this allows
us to search for minimal conditions (MCs) that guarantee
convergence to an attractor of interest. Not only are MCs
important to understand how to control complex network dynamics,
but they also allow us to quantify macro-level canalization therein.
From this, we get a measurable understanding of the robustness of
attractors of interest – the greater the canalization, the greater the
robustness to random perturbations – and, conversely, the
identification of critical node-states (enputs) in the network dynamics
to those attractors. We provided a measure of the capacity of these
critical nodes to control convergence to an attractor of interest
(enput power), and studied their robustness to disruptions. By
quantifying the ability of individual nodes to control attractor
behaviour, we can obtain a testable understanding of macro-level
canalization in the analysed biochemical network. Indeed, we can
uncover how robust phenotypic traits are (e.g. robustness of the
wild-type attractor), and which critical nodes must be acted upon
in order to disrupt phenotypic behaviour.
We exemplified our methodology with the well-known segment
polarity network model (in both the single-cell and the spatial
versions). Because this model has been extensively studied, we use it
to show that our analysis does not contradict any previous findings.
However, our analysis also allowed us to gain new knowledge about
its behaviour. From a better understanding of the size of its wild-
type attractor basin (larger than previously thought) to uncovering
new minimal conditions and critical nodes that control wild-type
behaviour. We also fully quantified micro- and macro-level
canalization in the model, and provided a complete map of its
canalization logic including dynamical modularity. Naturally, our
results pertain to this model; we do not claim that our results
characterize the real Drosophila segment polarity gene network.
However, our results, should they be found to deviate from
organism studies, can certainly be used to improve the current
model, and thus improve our understanding of Drosophila
development. Thus a key use of our methodology in systems
biology should be to help improve modelling accuracy. With the
methodology now tested on this model, in subsequent work we will
apply it to several automata network models of biochemical
regulation and signalling available in the systems biology literature.
The pathway modules we derived by inspection of the DCM for
the segment polarity network revealed a number of properties of
complex networks dynamics that deserve further study. For
instance, the dynamical sequence that occurs once each such
module is activated is independent of the temporal update scheme
utilized. Therefore, if the dynamics of a network is captured
exclusively by such modules, its intra-module behaviour will be
similar for both synchronous and asynchronous updating –
denoting a particular form of robustness to timing. We will
explore this property in future work, but as we showed here, the
dynamics of the single-cell version of the SPN model is very
(though not fully) controlled by only two pathway modules. This
explains why its dynamical behaviour is quite robust to timing
events as previously reported [38].
Research in cellular processes has provided a huge amount of
genomic, proteomic, and metabolomics data used to characterize
networks of biochemical reactions. All this information opens the
possibility of understanding complex regulation of intra- and inter-
cellular processes in time and space. However, this possibility is not
yet realized because we do not understand the dynamical
constraints that arise at the phenome (macro-) level from micro-
level interactions. One essential step towards reaching these
ambitious goals is to identify and understand the loci of control in
the dynamics of complex networks that make up living cells.
Towards this goal, we developed the new methodology presented
in this paper. Our methodology is applicable to any complex
network that can be modelled using binary state automata – and
easily extensible to multiple-state automata. We currently focus
only on biochemical regulation with the goal of understanding the
possible mechanisms of collective information processing that may
be at work in orchestrating cellular activity.
Supporting Information
Data S1
Glossary and mathematical notation.
(PDF)
Data S2
Details about the computation of wildcard and
two-symbol schemata.
(PDF)
Figure 21. Wild-type enput disruption in the SPN model. Each coordinate (x,y) in a given diagram (each corresponding to a tested enput)
contains a circle, depicting the proportion of trials that converged to attractor y when noise level x was used. Red circles mean that all trajectories
tested converged to y.
doi:10.1371/journal.pone.0055946.g021
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
32
March 2013 | Volume 8 | Issue 3 | e55946
Data S3
Details about the conversion of schemata into a
single threshold network.
(PDF)
Data S4
Algorithms for the computation of minimal
configurations.
(PDF)
Data S5
Further details concerning the minimal con-
figurations found for the segment polarity network
model.
(PDF)
Data S6
Basic notions of the inclusion/exclusion prin-
ciple.
(PDF)
Data S7
Minimal configurations for the segment polar-
ity network model obtained from biologically-plausible
seed configurations.
(CSV)
Data S8
Entire set of minimal configurations obtained
for the segment polarity network model.
(CSV)
Data S9
Minimal configurations for the segment polar-
ity network where no protein is on.
(CSV)
Data
S10
Minimal
configurations
for
the
segment
polarity network with the smallest number of nodes
that need to be specified in a Boolean state.
(CSV)
Data
S11
Minimal
configurations
for
the
segment
polarity network with the fewest number of on nodes.
(CSV)
Data
S12
Minimal
configurations
for
the
segment
polarity network with the largest number of on nodes.
(CSV)
Data S13
(Wildcard) minimal configurations for the
segment polarity network that were redescribed as two-
symbol schemata.
(CSV)
Data
S14
Minimal
configurations
for
the
segment
polarity
network
that
do
not
satisfy
wg4~1 _ en1~1 _ ci4~1.
(CSV)
Acknowledgments
We thank the FLAD Computational Biology Collaboratorium at the
Gulbenkian Institute of Science (Portugal) for hosting and providing
facilities used for this research. We also thank Indiana University for
providing access to its computing facilities. Finally, we are very grateful for
the generous and constructive comments we received from reviewers.
Author Contributions
Conceived and designed the presented methodology: MMP LMR.
Conceived and designed the experiments: MMP LMR. Performed the
experiments: MMP. Analyzed the data: MMP LMR. Wrote the paper:
MMP LMR.
References
1. Waddington CH (1942) Canalization of development and the inheritance of
acquired characters. Nature 150: 563–565.
2. Waddington CH (1957) The Strategy of the Genes: a Discussion of Some
Aspects of Theoretical Biology. London, UK: Allen & Unwin.
3. Conrad M (1990) The geometry of evolution. BioSystems 24: 61–81.
4. Fraser HB, Schadt EE (2010) The quantitative genetics of phenotypic
robustness. PLoS One 5: e8635.
5. Tusscher KHWJ, Hogeweg P (2009) The role of genome and gene regulatory
network canalization in the evolution of multi-trait polymorphisms and
sympatric speciation. BMC Evol Biol 9: 159.
6. Gibbon G (2009) Decanalization and the origin of complex disease. Nature
Reviews Genetics 10: 134–140.
7. Levy SF, Siegal ML (2008) Network hubs buffer environmental variation in
saccharomyces cerevisiae. PLoS Biol 6: e264.
8. Masel J, Maughan H (2007) Mutations leading to loss of sporulation ability in
bacillus subtilis are sufficiently frequent to favor genetic canalization. Genetics
175: 453–7.
9. Bergman A, Siegal ML (2003) Evolutionary capacitance as a general feature of
complex gene networks. Nature 424: 549–52.
10. Siegal M, Bergman A (2002) Waddington’s canalization revisited: Developmen-
tal stability and evolution. PNAS 99: 10528–10532.
11. Pigliucci M (2008) Is evolvability evolvable? Nature reviews Genetics 9: 75–82.
12. Kauffman S (1984) Emergent properties in random complex automata.
Physica D 10: 145–156.
13. Harris S, Sawhill B, Wuensche A, Kauffman S (2002) A model of transcriptional
regulatory networks based on biases in the observed regulation rules. Complexity
7: 23–40.
14. Kauffman S, Peterson C, Samuelsson B, Troein C (2003) Random boolean
network models and the yeast transcriptional network. PNAS 100: 14796–
14799.
15. Kauffman S, Peterson C, Samuelsson B, Troein C (2004) Genetic networks with
canalyzing boolean rules are always stable. PNAS.
16. Grefenstette J, Kim S, Kauffman S (2006) An analysis of the class of gene
regulatory functions implied by a biochemical model. BioSystems.
17. Raeymaekers L (2002) Dynamics of boolean networks controlled by biologically
meaningful functions. Journal of Theoretical Biology 218: 331–41.
18. Reichhardt CJO, Bassler K (2007) Canalization and symmetry in boolean
models for genetic regulatory networks. Physica A 40: 4339–4350.
19. Shmulevich I, La¨hdesma¨li H, Dougherty E, Jaakko A, Zhang W (2003) The role
of certain post classes in boolean network models of genetic networks. PNAS
100: 10734–10739.
20. Gat-Viks I, Shamir R (2003) Chain functions and scoring functions in genetic
networks. Bioinformatics 19 Suppl 1: i108–17.
21. Fortunato S (2010) Community detection in graphs. Physics Reports-Review
Section of Physics Letters 486: 75–174.
22. Kauffman S (1969) Homeostasis and differentiation in random genetic control
networks. Nature 224: 177–8.
23. Gershenson C (2004) Introduction to random boolean networks. In: Bedau M,
Husbands P, Hutton T, Kumar S, et al., editors, Workshop and Tutorial
Proceedings, Ninth International Conference on the Simulation and Synthesis of
Living Systems (ALife IX). 160–173.
24. Thomas R (1973) Boolean formalization of genetic control circuits. J Theor Biol
42: 563–85.
25. Thomas R, Thieffry D, Kaufman M (1995) Dynamical behaviour of biological
regulatory networks–i. biological role of feedback loops and practical use of the
concept of the loop-characteristic state. Bull Math Biol 57: 247–76.
26. Albert R, Othmer H (2003) The topology of the regulatory interactions predicts
the expression pattern of the segment polarity genes in drosophila melanogaster.
J Theor Biol 223: 1–18.
27. Alberts B, Bray D, Lewis J, Raff M, Roberts K, et al. (2003) Molecular Biology of
the Cell. Garland Publishing Group, 4th edition.
28. Wolpert L, Beddington R, Brockes J, Jessell T, Lawrence P, et al. (1998)
Principles of Development. London, UK: Current Biology Ltd.
29. Hooper J, Scott M (1992) The molecular genetic basis of positional information
in insect segments. In: Hennig W, editor, Early Embrionic Development of
Animals, Springer Verlag. 1–48.
30. von Dassow G, Meir E, Munro EM, Odell G (2000) The segment polarity
network is a robust developmental module. Nature 406: 188–192.
31. von Dassow G, Odell G (2002) Design and constraints of the Drosophila
segment polarity module: Robust spatial patterning emerges from intertwined
cell state switches. Journal of Experimental Zoology 294: 179–215.
32. Aldana M, Balleza E, Kauffman S, Resendiz O (2007) Robustness and
evolvability in genetic regulatory networks. J Theor Biol 245: 433–48.
33. Bornholdt S (2008) Boolean network models of cellular regulation: prospects and
limitations. J R Soc Interface 5 Suppl 1: S85–94.
34. Irons DJ (2009) Logical analysis of the budding yeast cell cycle. J Theor Biol 257:
543–59.
35. Alvarez-Buylla ER, Chaos A, Aldana M, Benitez M, Cortes-Poza Y, et al. (2008)
Floral morphogenesis: Stochastic explorations of a gene network epigenetic
landscape. Plos One 3.
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
33
March 2013 | Volume 8 | Issue 3 | e55946
36. Samaga R, Von Kamp A, Klamt S (2010) Computing combinatorial
intervention strategies and failure modes in signaling networks. J Comput Biol
17: 39–53.
37. Assmann SM, Albert R (2009) Discrete dynamic modeling with asynchronous
update. Methods in molecular biology 553: 207–225.
38. Chaves M, Albert R, Sontag ED (2005) Robustness and fragility of boolean
models for genetic regulatory networks. J Theor Biol 235: 431–449.
39. Marques-Pita M, Rocha LM (2011) Schema redescription in cellular automata:
Revisiting emergence in complex systems. In: Artificial Life (ALIFE), 2011 IEEE
Symposium on. 233–240. doi:10.1109/ALIFE.2011.5954670.
40. Quine WV (1955) A way to simplify truth functions. The American
Mathematical Monthly 62: 627–631.
41. Chandra A, Markowsky G (1978) On the number of prime implicants. Discrete
Mathematics : 7–11.
42. Blake A (1938) Canonical Expressions in Boolean Algebra. University of
Chicago Press.
43. Holland J, Holyoak K, Nisbett R, Thagard P (1986) Induction: Processes of
Inference, Learning and Discovery. MIT Press.
44. Karmiloff-Smith A (1992) Beyond Modularity: A Developmental Perspective on
Cognitive Science. MIT Press.
45. Klir GJ, Elias D (2002) Architecture of Systems Problem Solving. Da Capo
Press, Incorporated, 2 edition.
46. Valiant L (1984) A theory of the learnable. Communications of the ACM.
47. Wallace DAR (1998) Groups, rings, and fields. Berlin: Springer.
48. Shannon CE (1938) A symbolic analysis of relay and switching circuits. AIEE
Trans 57: 713–723.
49. McCluskey E (1956) Detection of group invariance or total symmetry of a
boolean function. Bell System Technical Journal 35: 1445–1453.
50. Kravets VN, Sakallah KA (2000) Generalized symmetries in boolean functions.
In: Sentovich E, editor, Proc. of Int. conf. on Computer Aided Design. IEEE,
526–532.
51. McCulloch W, Pitts W (1943) A logical calculus of ideas immanent in nervous
activity. Bulletin of Mathematical Biophysics 5: 115–133.
52. Willadsen K, Wiles J (2007) Robustness and state-space structure of boolean
gene regulatory models. J Theor Biol 249: 749–765.
53. Mitchell M (2006) Complex systems: Network thinking. Artificial Intelligence
170: 1194–1212.
54. Peak D, West JD, Messinger SM, Mott KA (2004) Evidence for complex,
collective dynamics and distributed emergent computation in plants. PNAS 101:
918–922.
55. Crutchfield J, Mitchell M (1995) The evolution of emergent computation. PNAS
92: 10742–10746.
56. Rocha LM, Hordijk W (2005) Material representations: From the genetic code
to the evolution of cellular automata. Artificial Life 11: 189–214.
57. Thieffry D, Huerta AM, Pe´rez-Rueda E, Collado-Vides J (1998) From specific
gene regulation to genomic networks: a global analysis of transcriptional
regulation in escherichia coli. Bioessays 20: 433–40.
58. Alexander RP, Kim PM, Emonet T, Gerstein MB (2009) Understanding
modularity in molecular networks requires dynamics. Sci Signal 2: pe44-.
59. Kolchinsky A, Rocha L (2011) Prediction and modularity in dynamical systems.
In: Proc. 11th conf. on the synthesis and simulation of living systems (ECAL
2011). MIT Press, 423–430.
60. Zallen JA, Wieschaus E (2004) Patterned gene expression directs bipolar planar
polarity in Drosophila. Developmental Cell 6: 343–355.
61. Lu X, Drocco J, Wieschaus EF (2010) Cell cycle regulation via inter-nuclear
communication during the early embryonic development of Drosophila
melanogaster. Cell cycle (Georgetown, Tex) 9: 2908–2910.
62. Mitchell M (1996) An Introduction to Genetic Algorithms. Cambridge, MA.:
MIT Press.
63. Bjorklund A, Husfeldt T (2006) Inclusion–exclusion algorithms for counting set
partitions. In: Foundations of Computer Science, 2006. FOCS ’06. 47th Annual
IEEE Symposium on. 575–582.
64. Helikar T, Konvalina J, Heidel J, Rogers JA (2008) Emergent decision-making
in biological signal transduction networks. PNAS 105: 1913–1918.
Canalization and Control in Automata Networks
PLOS ONE | www.plosone.org
34
March 2013 | Volume 8 | Issue 3 | e55946
|
23520449
|
HH_protein = ( hh )
EN_protein = ( en )
WG_protein = ( wg )
CIR = ( ( CI_protein AND ( ( ( PTC_protein ) ) ) ) AND NOT ( hh_external ) )
PTC_protein = ( ptc ) OR ( ( PTC_protein ) AND NOT ( hh_external ) )
ci = NOT ( ( EN_protein ) )
en = ( ( WG_external ) AND NOT ( SLP ) )
PH = ( PTC_protein AND ( ( ( hh_external ) ) ) )
wg = ( ( wg AND ( ( ( CIA OR SLP ) ) ) ) AND NOT ( CIR ) ) OR ( ( CIA AND ( ( ( SLP ) ) ) ) AND NOT ( CIR ) )
ptc = ( ( ( CIA ) AND NOT ( CIR ) ) AND NOT ( EN_protein ) )
hh = ( ( EN_protein ) AND NOT ( CIR ) )
SMO = ( ( hh_external ) ) OR NOT ( hh_external OR PTC_protein )
CI_protein = ( ci )
CIA = ( ( CI_protein ) AND NOT ( PTC_protein ) ) OR ( hh_external AND ( ( ( CI_protein ) ) ) )
|
A Comprehensive, Multi-Scale Dynamical Model of ErbB
Receptor Signal Transduction in Human Mammary
Epithelial Cells
Toma´sˇ Helikar1, Naomi Kochi1, Bryan Kowal2, Manjari Dimri3, Mayumi Naramura4,5,6, Srikumar M. Raja4,5,
Vimla Band4,5,6, Hamid Band4,5,6,7, Jim A. Rogers1,6*
1 Department of Mathematics, University of Nebraska at Omaha, Omaha, Nebraska, United States of America, 2 College of Information Technology, University of Nebraska
at Omaha, Omaha, Nebraska, United States of America, 3 George Washington University School of Medicine, Washington, D. C., United States of America, 4 The Eppley
Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, United States of America, 5 University of Nebraska Medical Center-Eppley Cancer Center, Omaha,
Nebraska, United States of America, 6 University of Nebraska Medical Center, Department of Genetics, Cell Biology and Anatomy, College of Medicine, Omaha, Nebraska,
United States of America, 7 University of Nebraska Medical Center, Departments of Biochemistry and Molecular Biology; Pathology and Microbiology; and Pharmacology
and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
Abstract
The non-receptor tyrosine kinase Src and receptor tyrosine kinase epidermal growth factor receptor (EGFR/ErbB1) have
been established as collaborators in cellular signaling and their combined dysregulation plays key roles in human cancers,
including breast cancer. In part due to the complexity of the biochemical network associated with the regulation of these
proteins as well as their cellular functions, the role of Src in EGFR regulation remains unclear. Herein we present a new
comprehensive, multi-scale dynamical model of ErbB receptor signal transduction in human mammary epithelial cells. This
model, constructed manually from published biochemical literature, consists of 245 nodes representing proteins and their
post-translational modifications sites, and over 1,000 biochemical interactions. Using computer simulations of the model,
we find it is able to reproduce a number of cellular phenomena. Furthermore, the model predicts that overexpression of Src
results in increased endocytosis of EGFR in the absence/low amount of the epidermal growth factor (EGF). Our subsequent
laboratory experiments also suggest increased internalization of EGFR upon Src overexpression under EGF-deprived
conditions, further supporting this model-generated hypothesis.
Citation: Helikar T, Kochi N, Kowal B, Dimri M, Naramura M, et al. (2013) A Comprehensive, Multi-Scale Dynamical Model of ErbB Receptor Signal Transduction in
Human Mammary Epithelial Cells. PLoS ONE 8(4): e61757. doi:10.1371/journal.pone.0061757
Editor: Nikos K. Karamanos, University of Patras, Greece
Received September 29, 2012; Accepted March 12, 2013; Published April 18, 2013
Copyright: 2013 Helikar et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work was supported by the National Institutes of Health (NIH) grants CA105489, CA87986, CA99163, CA116552 and NCI 5U01CA151806-02 and
Department of Defense grant W81WH-11-1-0167 to H.B; the NIH grants CA96844 and CA144027 and Department of Defense grants W81XWH-07-1-0351 and
W81XWH-11-1-0171 to V.B; Department of Defense grant W81 XWH-10-1-0740 to M.N.; the NCI Core Support Grant to UNMC-Eppley Cancer Center; by the
College of Arts and Sciences at the University of Nebraska at Omaha, the University of Nebraska Foundation, and Patrick J. Kerrigan and Donald F. Dillon
Foundations. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: jrogers@unomaha.edu
Introduction
EGF receptor (ErbB1) and other members of the ErbB family of
receptor tyrosine kinases (RTKs) play essential physiological roles
in development and maintenance of epithelial tissues by generat-
ing cell proliferation, survival, differentiation and migration signals
in response to specific ligands and via the stimulation of several
signaling pathways including PI3K/Akt, MAPK, Src, as well as
STAT pathways [1,2]. Activation of ErbB receptors is also linked
to the initiation and progression of human cancers. Thus,
elucidating signaling pathways that play critical roles in physio-
logical and oncogenic signaling by the ErbB family of receptors is
of substantial clinical significance [2–5]. Despite substantial
progress through experimental studies, in depth mechanistic
analyses of the signaling mechanisms of ErbB receptor family
have been quite challenging due to the multiple interactions
between members of the family, the number of associated effector
pathways, and the complexity of regulatory mechanisms [6]. In
addition to a multitude of positive signaling pathways triggered by
ErbB receptor activation, ErbB receptor signaling is also under
regulation by negative feedback mechanisms via receptor endo-
cytosis and recycling/degradation, and this mechanism is critical
for normal function [7]. The level of intricacy of the ErbB
signaling system is further multiplied by the fact that ErbB
signaling pathways are closely intertwined with a number of other
signaling pathways such as those downstream of integrins and G-
Protein-coupled Receptors [8]. Together, these complexities have
hampered our basic understanding of ErbB receptor signaling and
our ability to develop treatments for diseases, such as breast
cancer, lung cancer, gliomas and others, associated with aberrant
ErbB receptor signaling.
An example of the complex biology of ErbB receptor signaling
that is highly relevant to their role in oncogenesis involves the non-
receptor tyrosine kinase c-Src. The c-Src kinase is overexpressed
or hyperactive in a range of human tumors, including breast
cancer where as many as 70% cases have been reported with c-Src
overexpression along with EGFR/ErbB1 or ErbB2, leading to
conjectures of possible synergy between Src and the ErbBs in
PLOS ONE | www.plosone.org
1
April 2013 | Volume 8 | Issue 4 | e61757
breast cancer [9]. Indeed, in rodent fibroblasts [9,10] and more
importantly in untransformed human mammary epithelial cells
[11] the overexpression of c-Src promotes ErbB1/EGFR-depen-
dent oncogenic transformation. In particular, c-Src is a critical
component in the regulation of cell survival, proliferation as well as
migration, invasion and metastasis via the regulation of a number
of signaling pathways including PI3K/Akt, MAPK, as well as focal
adhesion kinase (FAK) [12]. However, the interconnectivity of
pathways associated with c-Src and the ErbB signaling has
hindered the determination of the mechanisms of ErbB-c-Src
synergy in cancer.
These difficulties represent an ideal example of the need for a
systems biology approach to ErbB receptor signaling. Because
ErbB1/EGFR has been extensively studied over the last several
decades, it is perhaps one of the best understood receptor tyrosine
kinase systems; this makes it a good candidate for computational
modeling [13]. Thus far, several EGFR-based computational
models have been created; these have been used in studies focusing
on receptor trafficking and endocytosis [14–17], ErbB dimeriza-
tion [18–20], as well as the relationships between physiological
responses and the receptor activation dynamics [21–23]. Several
modeling efforts have also been made to better understand the
signaling events downstream of EGFR [18,24–28]. In addition,
recent efforts also utilized a logical modeling approach to analyze
the topology and dynamics of an ErbB signaling network in
human liver cells [29], and to identify a potential new drug target,
c-MYC, in a model of ErbB receptor-mediated G1/S cell cycle
transition [30].
In this work, a new comprehensive, multi-scale logical model of
signal transduction in a human mammary epithelial cell (hMEC) is
presented. This large-scale dynamical model consists of 245
cellular components and about 1,100 biochemical interactions,
and encompasses all ErbB receptor family members, including
individual receptor phosphorylation sites, as well as integrin, G-
protein-coupled receptor, and stress signaling pathways. The
model was constructed manually by collecting and integrating
biochemical information from over 800 published papers. One of
the main advantages of logical models lies in their scalability; first,
they are based on qualitative information available for many
cellular components across many cell types and do not depend on
kinetic parameters (that are only sparsely available), and second,
simulations of logical models are relatively efficient [31,32],
making this approach appropriate for large systems.
Simulations of the model indicate a relatively accurate depiction
of the complexities of ErbB signaling by integrating a number of
other signaling pathways such as G-protein-coupled receptors
(GPCR) and integrins. Following its validation, the model was
used to generate predictions about the role of c-Src in EGFR
signaling, which were verified experimentally in the laboratory.
Finally, the model (including its governing logical expressions as
well as all annotations) is available on-line in The Cell Collective
software (www.thecellcollective.org; [32]) not only for download in
multiple open formats (e.g., SBML; [33]) , but also for live
simulations.
Results
1. The hMEC model
The hMEC model for EGFR signaling networks in a human
mammary epithelial cell was created by manually collecting
information on local biochemical interactions (e.g., protein-
protein) from the primary literature (using the same methods as
described in [34]). All interactions and logical expressions have
been cataloged and annotated, and are available in the Cell
Collective software [32]. In addition, all representing logical
expressions are available in Supporting Information S1. The
model contains a number of integrated signaling pathways at the
level of protein-protein as well as -post-translational (namely
phosphorylation) site interactions. These pathways include E-
cadherin, ErbB (1–4), ErbB1 (EGFR) endocytosis, G-protein-
coupled
Receptor,
integrin,
and
stress
signaling
pathways
(Figure 1). Detailed description of some of these follows below.
ErbB
receptors.
All
known
individual
ErbB
receptors
(ErbB1–4), as well as the major ErbB receptor dimers were
included in the model. Moreover, the hierarchy of the dimeriza-
tion process [35] was also captured via the logical functions
associated with each receptor node in the model (see the Model
Validation section). Furthermore, this model goes into an even
greater detail; all major (auto) phosphorylation sites of the ErbB
receptors were included. This allows for a range of mutational
studies at the phosphorylation site level in future studies; for
example, this multi-scale property of the mode will enable in silico
simulations and analyses of system-wide effects of all theoretically
possible combinations of virtual knock-out studies of the main
phosphorylation sites in a single receptor as well as across all ErbB
receptors.
EGFR endocytosis.
EGFR endocytosis as a potential mech-
anism of negative regulation through lysosomal degradation is
extremely important in relation to oncogenic signaling by ErbB
receptors [6]. Modeling receptor endocytosis is not trivial as the
same receptor can localize into different areas of the cell (e.g.,
clathrin-coated (CC) pits, CC vesicles, endosome, etc.), depending
on its stage of the endocytic trafficking. The way these multiple
localizations are handled using our modeling framework is to
represent EGFR with multiple nodes depicting the receptor in the
different locations during endocytic traffic. Specifically, the
locations included in the model are the following: the plasma
membrane, clathrin-coated pits, clathrin-coated vesicles, early
endosome, late endosome/multivesicular bodies (MVBs), and the
lysosome. The node representing EGFR in the lysosome denotes
the degradation process of EGFR. Because results from these
models do not attempt to predict exact measurements such as
concentrations of ligands used in laboratories, during a simulation,
the
activity
level
associated
with,
for
example,
the
node
representing EGFR in the lysosome corresponds to the degree of
EGFR being degraded in a semi-quantitative fashion. Further-
more, the type of ligand which activates EGFR has an effect on
whether the receptor is recycled back to cell surface or degraded
upon its internalization. To be able to simulate these effects in
addition to the nodes representing the different localizations of the
receptor during endocytosis, nodes depicting the receptor being
activated by different types of ligands (using EGF or TGFa as
prototypes) were added. Thus there is a node that represents
EGFR on the plasma membrane activated by EGF and a node for
EGFR activated by TGF-alpha. While these differentiations add
complexity to the model, they allow the visualization and study of
the dynamics of the model in greater detail.
Drugs and Antibodies.
Therapeutic agents that impact
ErbB receptor signaling and/or traffic, such as humanized anti-
ErbB2 monoclonal antibodies Herceptin and Pertuzumab, as well
as small molecular inhibitors are included in the presented model.
These components allow the simulation of mutations known to
cause cancer while at the same time introducing a number of
different levels and combinations of drugs and observing their
effects on the dynamics of the system.
In summary, the initial version of the new model of signal
transduction in human MEC comprises 240+ biological species
and 1,100+ biochemical interactions. The amount of detail
A Dynamical Model of Signal Transduction in hMEC
PLOS ONE | www.plosone.org
2
April 2013 | Volume 8 | Issue 4 | e61757
accomplished
by
including
individual
phosphorylation
sites,
various localizations, as well as dimers of ErbB receptors, and
the scale in terms of the number of the different pathways included
makes this model, to our best knowledge, the most comprehensive
dynamic model of signal transduction available to date.
2. Model Validation
The model was constructed using only information from the
primary literature about local interactions. In other words, during
the construction phase of the model there was no attempt to
determine the local interactions based on any other larger
phenotypes or phenomena. However, after the model was
completed, verification of the accuracy of the model involved
testing it for the ability to reproduce complex input-output
phenomena that have been observed in the laboratory. To do this,
The Cell Collective’s ‘‘Dynamical Analysis’’ simulation feature
was used [32]. This simulation component allows users to simulate
virtual cells under tens of thousands of cellular conditions, and
analyze and visualize the results in terms of input-output dose-
response curves that make it easy to determine whether the virtual
cell behaves as expected. The presented hMEC model was
interrogated to ensure that it is able to reproduce some of the
known global biological phenomena as previously observed
experimentally (Figure 2), including EGF-induced activation of
Figure 1. Graph representation of the model. The purpose of this graph is to visualize the complexity of the model, rather than to read the
individual interactions. This graph representation of the model was generated in Gephi (www.gephi.org).
doi:10.1371/journal.pone.0061757.g001
A Dynamical Model of Signal Transduction in hMEC
PLOS ONE | www.plosone.org
3
April 2013 | Volume 8 | Issue 4 | e61757
Akt and Erk, EGF-independent regulation of Erk via activated
Ras, integrin-dependent stimulation of Erk, Rac, and Cdc42, G-
protein Coupled Receptor activation of adenylyl cyclase, as well as
ErbB receptor dimerization hierarchy. Clearly, one cannot expect
that all phenomena will be replicated by the virtual cell due to the
fact that the model does not represent the entire cell. Hence, the
reproducibility of certain phenomena by the model only indicates
that the model is on the right track.
3. Use of the model to form laboratory-testable
predictions
Once a functional model of hMEC signaling was completed, we
used it to generate predictions about the system that could be
subsequently tested in the laboratory. In this work, the conjecture
of possible synergy between Src and the ErbBs in breast cancer
(described in the introduction) was explored using the model.
Specifically, the question examined was whether the internaliza-
tion of EGFR increases as a result of Src overexpression; this was
based on studies done in fibroblasts that showed increased EGFR
Figure 2. Validation of the MEC model against known cellular phenomena. A) EGF-dependent stimulation of survival signals via activation
of Akt. B) Activation of Erk by EGF [52]. Although a positive relationship between Erk and the ligand can be seen, the activation Erk by EGF seems to
exhibit a more complex dynamics than the ones seen in the other diagrams (i.e., the positive relationship is exhibited when EGF activity levels are 0–
40%). This is, however, not surprising given the complex interactions and cross-communication within the ErbB signaling family. C) Activating
mutations of known protooncogenes such as Ras result in growth factor-independent activation of Erk [53]. D) Erk dependency on signaling via
integrins by extracellular matrix (ECM) [54]. E and F) Activation of Rac and Cdc42 by ECM [55]. G) Positive relationship between Adenylyl Cyclase (AC)
and the G-Protein Coupled Receptor ligand alpha-s [56,57]. H) Hierarchy of ErbB receptor dimerization. The panel on the left represents a system with
EGFR expression alone and, thus, the formation of EGFR homodimers. The expression of ErbB2 (middle panel) results in the shift of the formation
(activity) from EGFR homodimers (EGFR-EGFR) to the formation of EGFR-ErbB2 heterodimers. Furthermore, the expression of ErbB 1–3 results in the
dominant formation of ErbB2–3 heterodimers [13,35]. Note that the references refer to classical, qualitative input-output relationships (not necessarily
quantitative dose–response curves), and the dose-response curves presented here are intended to demonstrate how the computational model
qualitatively reproduces the referenced input-output relationships over a range of stimulus signals.
doi:10.1371/journal.pone.0061757.g002
A Dynamical Model of Signal Transduction in hMEC
PLOS ONE | www.plosone.org
4
April 2013 | Volume 8 | Issue 4 | e61757
internalization in response to sub-saturating levels of EGF when
Src was overexpressed [36], but this phenomenon has not been
investigated in the more relevant MECs nor has it been tested at
low levels/in the absence of added EGF. To test whether the level of
Src activity affects EGFR endocytosis, the node representing c-Src
was
constitutively
activated
in
the
model,
using
the
Cell
Collective’s Dynamical Analysis feature. The model was then
simulated in The Cell Collective, and the activity levels of EGF-
activated EGFR homodimer on i) the plasma membrane, ii) in
clathrin-coated pits, iii) clathrin-coated vesicles, and iv) the
endosome were measured. The experiment was first conducted
with high levels of EGF to ensure that the model could predict the
known increase in internalization of EGFR under that condition.
The model was then simulated with low levels of EGF activity
(randomly ranging from 0–5%ON). As can be seen in Figure 3,
under both high and low levels of EGF, the overexpression of Src
(i.e., mutating Src to be constitutively-active) leads to the decrease
of the activity levels of the node representing EGFR homodimers
on the plasma membrane, while increasing the % ON levels of
nodes representing the receptor during the internalization process.
Thus the model indicates that Src overexpression will lead to
increased internalization of EGFR even in the absence (or low
levels) of EGF, a result that we assessed and confirmed
experimentally as discussed in the next section.
4. Verification of model-generated predictions
We sought to verify whether the predicted enhancement of the
intracellular (endocytosed) EGFR pools is seen in an actual human
MEC model in laboratory experiments. The overall levels of
ectopically expressed EGFR and Src in 76NTERT human MEC
lines retrovirally transuced with EGFR or EGFR plus Src (using
retroviral infection) [37] were assessed using Western blotting. As
is clear in Figure 4, increased Src and EGFR levels are observed in
the appropriate transductants. As expected, both the EGFR alone
transduced as well as the EGFR plus Src transduced cell lines
showed higher EGFR levels compared to the parent 76NTERT
cell line (Figure 4; results further explained in the caption).
Notably, the total levels of biochemically detected EGFR in the
EGFR plus Src transductant (lane 3) exceed the levels of EGFR
detectable in the EGFR alone transductant. In contrast, Fluores-
cence-activated cell sorting (FACS) based analysis that was
designed to selectively measure the levels of EGFR on the cell
surface (FACS analysis was done on live cells without permabiliz-
ing them; under these conditions, the staining antibody is excluded
from endocytosed pools of EGFR) demonstrated that, in the
absence of added EGF, there is dramatically reduced expression of
EGFR on the surface of MECs overexpressing Src (Figure 5, third
panel of the second row; indicated by the lower median
fluorescence channel values along the X-axis – MFI = 276)
compared to EGFR-only transduced MEC controls (Figure 5,
second panel of the second row, which expectedly shows a
markedly higher median channel value of 528 compared to112 in
untransduced control in the first panel). Thus, the prediction from
the hMEC model, that higher levels of Src introduced into MECs
will lead to a lower proportion of EGFR at the cell surface even in
the absence of EGF, i.e., EGFR is internalized, is fully verified by
experimental analyses of an actual mammary epithelial cell system.
Discussion
The critical roles of ErbB receptors in physiological processes
and the importance of their overexpression and/or hyperactivity
in the initiation and progression of cancers makes analyses of these
receptor tyrosine kinases extremely significant. EGFR is also the
most highly studied prototype of receptor tyrosine kinase signaling.
Therefore, predictive computational modeling of signaling down-
stream of these receptors in the context of positive and negative
loops and cross-talk with other receptor systems are likely to
greatly enhance our fundamental knowledge of cellular signaling
in health and disease. These models can also provide a more
informed platform for therapeutic targeting of aberrant ErbB
signaling in cancer to reduce treatment failures and to stem the
emergence of resistance. Here, we present a new and most
comprehensive computational model of ErbB receptor signaling in
a human mammary epithelial cell, use simulations to make a
specific prediction on the biological behavior of a hMEC under
experimental conditions that model the overexpression of EGFR
and Src as seen in human cancers, and then use laboratory
experiments to fully verify the prediction.
It is well-established that EGFR and c-Src tyrosine kinase are
co-overexpressed in human cancers, and experimental modeling
of their co-overexpression in untransformed hMECs in the
laboratory leads to their collaborative promotion of oncogenesis
[11]. Studies in such systems will be greatly enhanced by
generating hypotheses on the interactions of the two oncogenic
proteins that can be experimentally tested. As RTK signaling is
spatio-temporally regulated in part by the subcellular location of
activated receptors in different endocytic compartments which
serves as a critical determinant of the level and diversity of their
Figure 3. EGFR internalization effects by Src overexpression.
Experiments were conducted under conditions for which the expression
of ErbB2–4 was turned off and the virtual cell stimulated with EGF. A)
Stimulation of the virtual cell with low levels of EGF (0–5%). B) EGF was
introduced to the model in randomly selected high activity levels (60–
70% ON). * p,0.05 (Student’s t-test; n = 300; error bars represent the
standard error of the mean). Note: PM corresponds to EGFR
homodimers on the plasma membrane, CCP represents the homodimer
in clathrin-coated pits, CCV – clathrin-coated vesicles, and MVB – EGFR
homodimer in late endosome/multivescicular bodies.
doi:10.1371/journal.pone.0061757.g003
A Dynamical Model of Signal Transduction in hMEC
PLOS ONE | www.plosone.org
5
April 2013 | Volume 8 | Issue 4 | e61757
signaling outputs [37,38]. Therefore, we used the computational
model that we have developed to specifically address questions of
endocytic localization of EGFR under the influence of Src.
It is remarkable that the model could make an accurate de novo
prediction that the elevated Src activity will promote the
localization of EGFR in internal compartments of the endocytic
pathways even in the absence of added ligand. We used a cell
system in which we utilized an externally-introduced EGFR
(under a viral promoter) to test this prediction. Even though the
total levels of EGFR are substantially higher in the cell line that co-
overexpresses Src (Figure 4), the levels of EGFR on the cell surface
are dramatically reduced (Figure 5); thus, a vast majority of EGFR
in these cells is predicted to be in endocytic compartments even
without an added ligand. Under normal conditions, ligands such
as EGF cause rapid internalization of EGFR and its rapid
degradation in the lysosome while other ligands such as TGFalpha
or amphiregulin promote less degradation with more recycling
[39]. Notably, increased internal pools of EGFR are a feature of
oncogenic mutants of EGFR; we have recently shown that human
non-small cell lung cancer cell lines harboring EGFR mutants with
activating kinase domain mutations show increased levels of active
EGFR within intracellular endocytic compartments when cultured
without external EGF [40]. Remarkably, the internalized pool of
oncogenic EGFR co-localized with active Src [40] and the
interaction of mutant EGFR and Src was required for efficient
oncogenic transformation of fibroblasts [41]. Thus, our current
model-based prediction followed by experimental verification in a
relevant cell model in the laboratory should open a productive
lead to further experimentation towards understanding how Src
and EGFR cooperate in oncogenesis. Importantly, the verified
experimental findings can then be incorporated back into our
computational model to enhance its ability to make further
predictions and efforts along these lines are underway.
The use of computational models to generate experimentally
testable hypotheses in an information flow cycle from laboratories
to computational models and back to laboratories [42] is an
important dynamic that will define the future of computational
systems biology. However, the sheer size and complexity of
biochemical and biological processes poses a barrier for one
person or group to create and/or expand in an effective way large-
scale dynamical models of these systems. While the presented
model is one of the largest computational models created, it merely
represents a small fraction of the cell. Similar to Wikipedia and
open source software – both of which are centered around large
amounts of knowledge that could not have originated from one
single person or group, one way to create larger computational
models of biological process, ones that have the potential to
eventually lead to whole-cell models, is to engage the scientific
community in a collaborative fashion. Hence, the presented
hMEC model has been made available in The Cell Collective
software which was designed precisely to enable such a collabo-
rative and collective approach to systems biology [32]. The model
is available to the entire scientific community via the software for
further expansion, refinements, as well as simulations and analyses.
The user-friendly interface of the software allows users to make
changes to the model without any need to enter complex
mathematical equations or computer code, making it accessible
to experimental scientists who have the most intimate knowledge
of the local data to improve and grow this model (and others
available in the software platform; e.g., [30,43,44]).
Materials and Methods
1. Model construction via The Cell Collective
The presented model is based on a common qualitative
(discrete) modeling technique where the regulatory mechanism
of each node is described by a Boolean expression (for more
comprehensive information on Boolean modeling see for example
[45,46]). The construction of the model was accomplished using
The Cell Collective (www.thecellcollective.org; [32]), a collabora-
tive modeling platform for large-scale biological systems. The
platform allows users to construct and simulate large-scale
computational models of various biological processes based on
qualitative interaction information using the platform’s Bio-Logic
Builder which converts the entered qualitative biochemical
information into Boolean expressions in the background [47].
(Though the Boolean expressions for any model created in the
platform can be downloaded from the website.) This non-technical
creation and representation of the individual interactions in the
model make it especially easy for experimental biologists to
contribute to the creation of the model without the need for
training in the underlying mathematical formalisms. The model
has been exported and is also available as part of this manuscript
in a SBML format for qualitative models (Supporting Information
S2).
The Cell Collective’s Knowledge Base component was also used
to catalog and annotate all biochemical/biological information for
signaling in hMECs as mined from the primary literature. Each
Figure 4. Src overexpression does not affect total EGFR levels.
Parental 76N-TERT cells or its EGFR or EGFR + Src transductants were
EGF deprived by culture in the EGF-deficient D3 medium for 48 hrs and
cell lysates were prepared. 50 mg aliquots of cell lysate protein were run
on an 8% SDS PAGE gel and immunoblotted with anti-EGFR (top panel)
or anti-Src (middle panel). Membrane was re-probed with anti-beta
actin (bottom panel) to ensure equal loading.
doi:10.1371/journal.pone.0061757.g004
A Dynamical Model of Signal Transduction in hMEC
PLOS ONE | www.plosone.org
6
April 2013 | Volume 8 | Issue 4 | e61757
model species in the Knowledge Base has its own wiki-like page
where information on the individual interactions is stored,
including references.
2. Model Simulations and Analysis
In addition to the model building and cataloging process, The
Cell Collective platform was also used to perform all computa-
tional simulations of the hMEC model. While the dynamical
model is based on a discrete (i.e., Boolean) formalism, as can be
seen in the results, the simulation input and output data are
continuous. This was accomplished by converting the digital
output of the model simulations to % activity (% ON) which
ranges (for each model component) from 0 to 100 [31,34]. It is
important to note that the % ON doesn’t directly correspond to
the biological concentration or any other measurable property,
rather the % ON provides a semi-quantitative measure to describe
the relative activity level of a particular protein. As such, the model
output (species activity levels) is compared to previously published
experimental findings as well as the experimental results presented
herein by assessing the directionality of the changes (up-/down-
regulation) of species activity relative to the wild-type. All
simulations were conducted using a biologically relevant initial
condition as discussed in [34]; this condition is also accessible via
The Cell Collective software. All in silico experiments were
performed
under
external
conditions
(Table
1)
that
were
optimized for the particular experiment [34].
3. Establishment of mammary epithelial cell lines
overexpressing EGFR or EGFR plus Src
Human telomerase reverse transcriptase (TERT) immortalized
76N normal human MEC line 76N-TERT has been previously
described [48,49]. These cells were cultured in DFCI-1 medium
and the human EGFR or EGFR plus c-Src were overexpressed in
these cells using retroviral infection as described previously [50].
4. Assessment of Src overexpression and EGFR levels
using Western blotting
Parental 76NTERT cell line or its EGFR or EGFR + Src
transductants were growth factor deprived for 48 h (with medium
change each day) in EGF-deficient D3 medium (DFCI-1 medium
lacking insulin, hydrocortisone, EGF and bovine pituitary extract)
Figure 5. Surface expression of EGFR is reduced in Src-transfected cells. FACS analysis of the cell surface expression of EGFR in MEC
transductants shows (in row 2) that Src overexpression leads to a reduction in the level of EGFR at the cell surface despite higher total EGFR levels
detected biochemically (shown in Figure 3). Cells were EGF-deprived by culture in D3 medium for 48 h, after which single cell suspensions were
prepared with trypsin/EDTA. Live cells were stained with isotype-matched control monoclonal antibodies or with anti-EGFR monoclonal antibody for
1 hr.., washed and incubated with the secondary antibody (PE-conjugated) for 45 min., and analyzed by FACS to determine cell surface levels of
EGFR. Numbers in the right top corner of each FACS panel indicate median fluorescence channel intensity (higher numbers indicating higher levels
and vice versa). Initial experiments (not shown) established the specificity of antibodies.
doi:10.1371/journal.pone.0061757.g005
A Dynamical Model of Signal Transduction in hMEC
PLOS ONE | www.plosone.org
7
April 2013 | Volume 8 | Issue 4 | e61757
[51], Cell lysates were prepared in a Triton X-100-based lysis
buffer, and 50 mg aliquots of cell lysate protein were run on an 8%
SDS PAGE gel and immunoblotted with anti-EGFR, anti-Src and
anti-beta actin antibodies, as described [50].
5. Assessment of surface expression of EGFR
Cells were cultured under EGF-deprivation conditions as above
for 48 h, and single cell suspensions were prepared by releasing
cells from tissue culture plates with trypsin/EDTA. Live cells were
stained on ice with isotype-matched control monoclonal antibodies
or with anti-EGFR monoclonal antibody (clone 528; ATCC) for
1 h, washed and incubated with the secondary antibody (PE-
conjugated anti-mouse IgG) for 45 min followed by FACS analysis
(using a FACSCalibur instrument) to determine the relative cell
surface EGFR levels.
Supporting Information
Supporting Information S1
A comprehensive, multi-scale
dynamical model of ErbB receptor signal transduction in human
mammary epithelial cells
(DOC)
Supporting Information S2
SBML model representation of
signal transduction epithelial cells
(SBML)
Author Contributions
Conceived and designed the experiments: TH JAR HB MN SMR VB
MD. Performed the experiments: TH NK JAR HB MN SMR VB MD.
Analyzed the data: TH JAR HB MN SMR VB MD. Contributed
reagents/materials/analysis tools: TH BK JAR HB MN SMR VB MD.
Wrote the paper: TH JAR HB.
References
1. Hynes NE, Lane HA (2005) ERBB receptors and cancer: the complexity of
targeted inhibitors. Nature reviews Cancer 5: 341–354. doi:10.1038/nrc1609.
2. Hynes NE, MacDonald G (2009) ErbB receptors and signaling pathways in
cancer. Curr Opin Cell Biol 21: 177–184.
3. Stern DF (2000) Tyrosine kinase signalling in breast cancer: ErbB family
receptor tyrosine kinases. Breast Cancer Res 2: 176–183.
4. Yarden Y (2001) Biology of HER2 and its importance in breast cancer.
Oncology 61 Suppl 2: 1–13.
5. Holbro T, Beerli RR, Maurer F, Koziczak M, Barbas CF 3rd, et al. (2003) The
ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires
ErbB3 to drive breast tumor cell proliferation. Proc Natl Acad Sci U S A 100:
8933–8938.
6. Yarden Y, Sliwkowski MX (2001) Untangling the ErbB signalling network. Nat
Rev Mol Cell Biol 2: 127–137.
7. Sorkin A, Von Zastrow M (2009) Endocytosis and signalling: intertwining
molecular networks. Nat Rev Mol Cell Biol 10: 609–622.
8. Wertheimer E, Gutierrez-Uzquiza A, Rosemblit C, Lopez-Haber C, Sosa MS,
et al. (2012) Rac signaling in breast cancer: a tale of GEFs and GAPs. Cellular
signalling 24: 353–362. doi:10.1016/j.cellsig.2011.08.011.
9. Biscardi JS, Ishizawar RC, Silva CM, Parsons SJ (2000) Tyrosine kinase
signalling in breast cancer: epidermal growth factor receptor and c-Src
interactions in breast cancer. Breast Cancer Res 2: 203–210.
10. Maa MC, Leu TH, McCarley DJ, Schatzman RC, Parsons SJ (1995)
Potentiation of epidermal growth factor receptor-mediated oncogenesis by c-
Src: implications for the etiology of multiple human cancers. Proceedings of the
National Academy of Sciences of the United States of America 92: 6981–6985.
11. Dimri M, Naramura M, Duan L, Chen J, Ortega-Cava C, et al. (2007) Modeling
breast cancer-associated c-Src and EGFR overexpression in human MECs: c-
Src and EGFR cooperatively promote aberrant three-dimensional acinar
structure and invasive behavior. Cancer research 67: 4164–4172.
doi:10.1158/0008-5472.CAN-06-2580.
12. Wheeler DL, Iida M, Dunn EF (2009) The role of Src in solid tumors.
Oncologist 14: 667–678.
13. Wiley HS, Shvartsman SY, Lauffenburger DA (2003) Computational modeling
of the EGF-receptor system: a paradigm for systems biology. Trends Cell Biol
13: 43–50.
14. Shankaran H, Wiley HS, Resat H (2007) Receptor downregulation and
desensitization enhance the information processing ability of signalling receptors.
BMC Syst Biol 1: 48.
15. Gex-Fabry M, DeLisi C (1984) Receptor-mediated endocytosis: a model and its
implications for experimental analysis. Am J Physiol 247: R768–79.
16. Wiley HS, Cunningham DD (1981) A steady state model for analyzing the
cellular binding, internalization and degradation of polypeptide ligands. Cell 25:
433–440.
17. Wiley HS, Cunningham DD (1982) The endocytotic rate constant. A cellular
parameter for quantitating receptor-mediated endocytosis. J Biol Chem 257:
4222–4229.
18. Hendriks BS, Orr G, Wells A, Wiley HS, Lauffenburger DA (2005) Parsing
ERK activation reveals quantitatively equivalent contributions from epidermal
Table 1. External simulation conditions.
Stimulus/Simulation
Experiment
Fig. 2A
Fig. 2B
Fig. 2C
Fig. 2D Fig. 2E
Fig. 2F
Fig. 2G Fig. 2H
Fig. 3A
Fig. 3B
alpha_1213L
40–60
72
72
60–70
5–65
70–80
36
26
20–30
20–30
alpha_iL
0–10
11
11
70–80
30–80
40–50
69
97
90
100
alpha_qL
0–10
37
37
10–20
50–100
90–100
77
4
1
5
alpha_sL
40–60
48
48
10–20
55–95
50–60
0–100
55
50
60
ECM
0–100
88
88
0–100
0–100
0–100
99
94
90
100
EGF
0–100
0–100
0–100
0
0–100
1–10
10
90–100
0–5
60–70
EGFR_Contr
100
100
100
100
100
100
100
90–100
100
100
ErbB2_Contr
0
0
0
0
0
0
0
0/90–100/90–100
0
0
ErbB2Deg_Contr
0
0
0
0
0
0
0
0/25–35/25–35
0
0
ErbB3_Contr
0
0
0
0
0
0
0
0/0/90–100
0
0
ExtPump
40–60
12
12
1–10
80–100
90–100
94
76
70–80
70–80
IL1_TNF
2
2
2
2
2
2
2
2
2
2
Stress
2
2
2
2
2
2
2
2
2
2
All values represent (%ON) activity levels. Stimuli not used in these experiments were set to 0 and are not listed in the table. Note that while some external conditions
consist of ranges and others of specific values, we find that most experiments are not sensitive to specific values. Using the values provided in this table, all simulated
experiments of the model can be reproduced in The Cell Collective software.
doi:10.1371/journal.pone.0061757.t001
A Dynamical Model of Signal Transduction in hMEC
PLOS ONE | www.plosone.org
8
April 2013 | Volume 8 | Issue 4 | e61757
growth factor receptor and HER2 in human mammary epithelial cells. J Biol
Chem 280: 6157–6169.
19. Hendriks BS, Opresko LK, Wiley HS, Lauffenburger D (2003) Coregulation of
epidermal growth factor receptor/human epidermal growth factor receptor 2
(HER2) levels and locations: quantitative analysis of HER2 overexpression
effects. Cancer Res 63: 1130–1137.
20. Shankaran H, Wiley HS, Resat H (2006) Modeling the effects of HER/ErbB1-3
coexpression on receptor dimerization and biological response. Biophys J 90:
3993–4009.
21. Knauer DJ, Wiley HS, Cunningham DD (1984) Relationship between
epidermal growth factor receptor occupancy and mitogenic response. Quanti-
tative analysis using a steady state model system. J Biol Chem 259: 5623–5631.
22. Starbuck C, Lauffenburger DA (1992) Mathematical model for the effects of
epidermal growth factor receptor trafficking dynamics on fibroblast proliferation
responses. Biotechnol Prog 8: 132–143.
23. Waters CM, Overholser KA, Sorkin A, Carpenter G (1992) Analysis of the
influences of the E5 transforming protein on kinetic parameters of epidermal
growth factor binding and metabolism. J Cell Physiol 152: 253–263.
24. Bhalla US, Iyengar R (1999) Emergent properties of networks of biological
signaling pathways. Science 283: 381–387.
25. Kholodenko BN, Demin O V, Moehren G, Hoek JB (1999) Quantification of
short term signaling by the epidermal growth factor receptor. J Biol Chem 274:
30169–30181.
26. Haugh JM, Huang AC, Wiley HS, Wells A, Lauffenburger DA (1999)
Internalized epidermal growth factor receptors participate in the activation of
p21(ras) in fibroblasts. J Biol Chem 274: 34350–34360.
27. Schoeberl B, Eichler-Jonsson C, Gilles ED, Mu¨ller G (2002) Computational
modeling of the dynamics of the MAP kinase cascade activated by surface and
internalized EGF receptors. Nat Biotechnol 20: 370–375.
28. Wang DYQ, Cardelli L, Phillips A, Piterman N, Fisher J (2009) Computational
modeling of the EGFR network elucidates control mechanisms regulating signal
dynamics. BMC Syst Biol 3: 118.
29. Samaga R, Saez-Rodriguez J, Alexopoulos LG, Sorger PK, Klamt S (2009) The
logic of EGFR/ErbB signaling: theoretical properties and analysis of high-
throughput data. PLoS Comput Biol 5: e1000438.
30. Sahin O, Fro¨hlich H, Lo¨bke C, Korf U, Burmester S, et al. (2009) Modeling
ERBB receptor-regulated G1/S transition to find novel targets for de novo
trastuzumab resistance. BMC Syst Biol 3: 1.
31. Helikar T, Rogers JA (2009) ChemChains: a platform for simulation and
analysis of biochemical networks aimed to laboratory scientists. BMC Syst Biol
3: 58.
32. Helikar T, Kowal B, McClenathan S, Bruckner M, Rowley T, et al. (2012) The
Cell Collective: Toward an open and collaborative approach to systems biology.
BMC systems biology 6: 96. doi:10.1186/1752-0509-6-96.
33. Finney A, Hucka M (2003) Systems biology markup language: Level 2 and
beyond. Biochemical Society transactions 31: 1472–1473. doi:10.1042/.
34. Helikar T, Konvalina J, Heidel J, Rogers JA (2008) Emergent decision-making
in biological signal transduction networks. Proc Natl Acad Sci U S A 105: 1913–
1918.
35. Tzahar E, Waterman H, Chen X, Levkowitz G, Karunagaran D, et al. (1996) A
hierarchical network of interreceptor interactions determines signal transduction
by Neu differentiation factor/neuregulin and epidermal growth factor. Mol Cell
Biol 16: 5276–5287.
36. Ware MF, Tice DA, Parsons SJ, Lauffenburger DA (1997) Overexpression of
cellular Src in fibroblasts enhances endocytic internalization of epidermal growth
factor receptor. J Biol Chem 272: 30185–30190.
37. Sorkin A, Goh LK (2008) Endocytosis and intracellular trafficking of ErbBs.
Experimental cell research 314: 3093–3106. doi:10.1016/j.yexcr.2008.08.013.
38. Roepstorff K, Grøvdal L, Grandal M, Lerdrup M, Van Deurs B (2008)
Endocytic downregulation of ErbB receptors: mechanisms and relevance in
cancer. Histochemistry and cell biology 129: 563–578. doi:10.1007/s00418-008-
0401-3.
39. Zwang Y, Yarden Y (2009) Systems biology of growth factor-induced receptor
endocytosis. Traffic (Copenhagen, Denmark) 10: 349–363. doi:10.1111/j.1600-
0854.2008.00870.x.
40. Chung BM, Raja SM, Clubb RJ, Tu C, George M, et al. (2009) Aberrant
trafficking of NSCLC-associated EGFR mutants through the endocytic recycling
pathway promotes interaction with Src. BMC cell biology 10: 84. doi:10.1186/
1471-2121-10-84.
41. Chung BM, Dimri M, George M, Reddi AL, Chen G, et al. (2009) The role of
cooperativity with Src in oncogenic transformation mediated by non-small cell
lung cancer-associated EGF receptor mutants. Oncogene 28: 1821–1832.
doi:10.1038/onc.2009.31.
42. Di Ventura B, Lemerle C, Michalodimitrakis K, Serrano L (2006) From in vivo
to in silico biology and back. Nature 443: 527–533.
43. Todd RG, Helikar T (2012) Ergodic sets as cell phenotype of budding yeast cell
cycle. PloS one 7: e45780. doi:10.1371/journal.pone.0045780.
44. Madrahimov A, Helikar T, Kowal B, Lu G, Rogers J (2012) Dynamics of
Influenza Virus and Human Host Interactions During Infection and Replication
Cycle. Bulletin of mathematical biology In press. doi:10.1007/s11538-012-9777-
2.
45. Morris MK, Saez-Rodriguez J, Sorger PK, Lauffenburger DA (2010) Logic-
based models for the analysis of cell signaling networks. Biochemistry 49: 3216–
3224. doi:10.1021/bi902202q.
46. Helikar T, Kochi N, Konvalina J, Rogers JA (2011) Boolean Modeling of
Biochemical Networks. The Open Bioinformatics Journal 4: 16–25.
47. Helikar T, Kowal B, Madrahimov A, Shrestha M, Pedersen J, et al. (2012) Bio-
logic builder: a non-technical tool for building dynamical, qualitative models.
PloS one 7: e46417. doi:10.1371/journal.pone.0046417.
48. Zhao X, Malhotra GK, Lele SM, Lele MS, West WW, et al. (2010) Telomerase-
immortalized human mammary stem/progenitor cells with ability to self-renew
and differentiate. Proceedings of the National Academy of Sciences of the
United States of America 107: 14146–14151. doi:10.1073/pnas.1009030107.
49. Ratsch SB, Gao Q, Srinivasan S, Wazer DE, Band V (2001) Multiple genetic
changes are required for efficient immortalization of different subtypes of normal
human mammary epithelial cells. Radiation research 155: 143–150.
50. Dimri M, Naramura M, Duan L, Chen J, Ortega-Cava C, et al. (2007) Modeling
breast cancer-associated c-Src and EGFR overexpression in human MECs: c-
Src and EGFR cooperatively promote aberrant three-dimensional acinar
structure and invasive behavior. Cancer research 67: 4164–4172.
doi:10.1158/0008-5472.CAN-06-2580.
51. Band V, Sager R (1989) Distinctive traits of normal and tumor-derived human
mammary epithelial cells expressed in a medium that supports long-term growth
of both cell types. Proceedings of the National Academy of Sciences of the
United States of America 86: 1249–1253.
52. Roux PP, Blenis J (2004) ERK and p38 MAPK-activated protein kinases: a
family of protein kinases with diverse biological functions. Microbiology and
molecular
biology
reviews:
MMBR
68:
320–344.
doi:10.1128/
MMBR.68.2.320-344.2004.
53. Sridhar SS, Hedley D, Siu LL (2005) Raf kinase as a target for anticancer
therapeutics. Molecular cancer therapeutics 4: 677–685. doi:10.1158/1535-
7163.MCT-04-0297.
54. Edin ML, Juliano RL (2005) Raf-1 serine 338 phosphorylation plays a key role in
adhesion-dependent activation of extracellular signal-regulated kinase by
epidermal growth factor. Molecular and cellular biology 25: 4466–4475.
doi:10.1128/MCB.25.11.4466-4475.2005.
55. Price LS, Leng J, Schwartz MA, Bokoch GM (1998) Activation of Rac and
Cdc42 by integrins mediates cell spreading. Molecular biology of the cell 9:
1863–1871.
56. Selbie LA, Hill SJ (1998) G protein-coupled-receptor cross-talk: the fine-tuning
of multiple receptor-signalling pathways. Trends in pharmacological sciences 19:
87–93.
57. Milligan G, White JH (2001) Protein-protein interactions at G-protein-coupled
receptors. Trends in pharmacological sciences 22: 513–518.
A Dynamical Model of Signal Transduction in hMEC
PLOS ONE | www.plosone.org
9
April 2013 | Volume 8 | Issue 4 | e61757
|
23637902
|
Grb2 = ( EGFR_Y1068 ) OR ( EGFR_Y1086 ) OR ( ErbB2_Y1139 ) OR ( Src AND ( ( ( Fak ) ) ) ) OR ( Shc )
ILK = ( PIP3_345 )
PIP2_45 = ( PTEN AND ( ( ( PIP3_345 ) ) ) ) OR ( PI4K AND ( ( ( PI5K ) ) ) ) OR ( ( PIP2_45 ) AND NOT ( PI3K AND ( ( ( PIP2_45 ) ) ) ) )
PKC = ( ( AA AND ( ( ( PKC_primed ) ) AND ( ( Ca ) ) ) ) AND NOT ( Trx AND ( ( ( PKC ) ) ) ) ) OR ( ( PKC AND ( ( ( NOT PP2A ) ) AND ( ( NOT Trx ) ) ) ) AND NOT ( Trx AND ( ( ( PKC ) ) ) ) ) OR ( ( DAG AND ( ( ( Ca ) ) AND ( ( PKC_primed ) ) ) ) AND NOT ( Trx AND ( ( ( PKC ) ) ) ) )
Cdc42 = ( ( Cdc42 AND ( ( ( NOT p190RhoGAP AND NOT Graf AND NOT RalBP1 ) ) AND ( ( IQGAP1 ) ) ) ) AND NOT ( RhoGDI AND ( ( ( Src ) ) ) ) ) OR ( ( Pix_Cool AND ( ( ( PAK AND Gbg_i ) AND ( ( ( NOT Rac ) ) ) ) OR ( ( Cdc42 ) ) ) ) AND NOT ( RhoGDI AND ( ( ( Src ) ) ) ) )
ErbB2_Y1221_22 = ( EGFR_ErbB2 ) OR ( ErbB2_ErbB3 ) OR ( ErbB2_ErbB4 )
ARF = ( ARNO AND ( ( ( NOT PIP2_45 ) ) ) ) OR ( ARF AND ( ( ( NOT PIP2_45 ) ) ) ) OR ( PIP3_345 AND ( ( ( NOT PIP2_45 ) ) ) ) OR ( PIP2_45 AND ( ( ( PIP3_345 OR ARNO ) AND ( ( ( NOT ARF ) ) ) ) ) )
Talin = ( PIP2_45 AND ( ( ( NOT Talin ) ) ) ) OR ( Talin AND ( ( ( NOT Src ) ) ) )
Mekk4 = ( Rac ) OR ( Cdc42 )
ErbB4_Y1242 = ( ErbB4_ErbB4 ) OR ( ErbB3_ErbB4 ) OR ( EGFR_ErbB4 ) OR ( ErbB2_ErbB4 )
RKIP = ( PKC )
Ga_1213 = ( Ga_1213 AND ( ( ( NOT p115RhoGEF ) ) AND ( ( Gbg_1213 ) ) ) ) OR ( alpha_1213R AND ( ( ( NOT Ga_1213 AND NOT Gbg_1213 ) ) ) )
CaMKK = ( CaM )
Cbl_RTK = ( ( ( Grb2 AND ( ( ( Src ) ) ) ) AND NOT ( EGFR_T654 ) ) AND NOT ( CIN85 AND ( ( ( Spry2 ) ) ) ) ) OR ( ( ( EGFR_Y1045 AND ( ( ( Src ) ) ) ) AND NOT ( EGFR_T654 ) ) AND NOT ( CIN85 AND ( ( ( Spry2 ) ) ) ) )
RGS = ( CaM AND ( ( ( PIP3_345 ) ) ) )
EGFR_Y1148 = ( EGFR_ErbB3 ) OR ( EGFR_ErbB2 ) OR ( EGFR_ErbB4 ) OR ( EGFR_EGFR ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
Tiam = ( PKC AND ( ( ( Ras OR Rap1 OR PIP2_45 ) ) AND ( ( PIP3_345 OR PIP2_34 ) ) ) ) OR ( Src AND ( ( ( Ras OR Rap1 OR PIP2_45 ) ) AND ( ( PIP3_345 OR PIP2_34 ) ) ) ) OR ( CaMK AND ( ( ( PIP3_345 OR PIP2_34 ) ) AND ( ( Ras OR Rap1 OR PIP2_45 ) ) ) )
Hsp90 = ( ( EGFR_ErbB2 ) AND NOT ( CHIP AND ( ( ( Hsp90 ) ) ) ) ) OR ( ( ErbB2_ErbB3 ) AND NOT ( CHIP AND ( ( ( Hsp90 ) ) ) ) ) OR ( ( ErbB2_ErbB4 ) AND NOT ( CHIP AND ( ( ( Hsp90 ) ) ) ) )
ErbB3_ErbB4 = ( ( NRG AND ( ( ( ErbB4_Free AND ErbB3_Free ) ) ) ) AND NOT ( ErbB2_Free ) )
PA = ( PLD )
RhoK = ( Rho )
GAK = ( GAK AND ( ( ( EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_EGF_CCV ) ) ) ) OR ( EGFR_EGFR_EGF_CCP AND ( ( ( Clathrin OR Dynamin OR AP2 ) ) ) ) OR ( EGFR_EGFR_TGFa_CCP AND ( ( ( Clathrin OR Dynamin OR AP2 ) ) ) )
EGFR_EGFR_EGF_Lysosome = ( EGFR_EGFR_EGF_MVB AND ( ( ( VPS4 OR Eps15 ) ) AND ( ( Rab7 ) ) AND ( ( Alix ) ) ) ) OR ( ESCRT_III AND ( ( ( VPS4 OR Eps15 ) ) AND ( ( Rab7 ) ) AND ( ( Alix ) ) ) )
EGFR_ErbB2 = ( Pertuzumab AND ( ( ( EGFR_ErbB2 ) ) ) ) OR ( ( ( EGFR_ErbB2 ) AND NOT ( Trastuzumab ) ) AND NOT ( ErbB2Deg_Contr AND ( ( ( EGFR_ErbB2 ) ) ) ) ) OR ( ( EGF AND ( ( ( NOT NRG OR NOT ErbB3_Free ) ) AND ( ( NOT EGFR_ErbB2 ) ) AND ( ( NOT EGFR_ErbB2 ) ) AND ( ( ErbB2_Free AND EGFR_Free ) ) AND ( ( NOT EGFR_T654 ) ) ) ) AND NOT ( Pertuzumab ) )
EGFR_Y1173 = ( EGFR_ErbB3 ) OR ( EGFR_ErbB2 ) OR ( EGFR_ErbB4 ) OR ( EGFR_EGFR ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
alpha_catenin = ( B_catenin )
SAPK = ( ( ( MKK7 ) AND NOT ( MKPs AND ( ( ( SAPK ) ) ) ) ) AND NOT ( PP2A AND ( ( ( SAPK ) ) ) ) ) OR ( ( ( Sek1 ) AND NOT ( MKPs AND ( ( ( SAPK ) ) ) ) ) AND NOT ( PP2A AND ( ( ( SAPK ) ) ) ) )
ErbB2_ErbB3 = ( ( ErbB2_ErbB3 ) AND NOT ( ErbB2_Lysosome AND ( ( ( ErbB2_ErbB3 ) ) ) ) ) OR ( ( ( NRG AND ( ( ( ErbB2_Free AND ErbB3_Free ) ) ) ) AND NOT ( Pertuzumab ) ) AND NOT ( ErbB2_Lysosome AND ( ( ( ErbB2_ErbB3 ) ) ) ) )
Cortactin = ( Fer ) OR ( Src ) OR ( Actin ) OR ( Rac ) OR ( Dynamin ) OR ( Hip1R ) OR ( PAK ) OR ( Erk )
Hip1R = ( Clathrin ) OR ( CIN85 )
PIP2_34 = ( PIP2_34 AND ( ( ( NOT PI5K ) ) AND ( ( NOT PTEN ) ) ) ) OR ( PI4K AND ( ( ( NOT PIP2_34 ) ) AND ( ( PI3K ) ) ) )
EGFR_ErbB4 = ( EGF AND ( ( ( NOT EGFR_T654 ) ) AND ( ( NOT ErbB3_Free ) ) AND ( ( ErbB4_Free ) ) AND ( ( NOT ErbB2_Free ) ) AND ( ( NRG ) ) AND ( ( EGFR_Free ) ) ) )
p120RasGAP = ( ( ( EGFR_Y992 ) AND NOT ( Src ) ) AND NOT ( Fak ) ) OR ( ( ( PIP3_345 ) AND NOT ( Src ) ) AND NOT ( Fak ) ) OR ( ( ( PIP2_45 ) AND NOT ( Src ) ) AND NOT ( Fak ) ) OR ( ( ( PIP2_34 ) AND NOT ( Src ) ) AND NOT ( Fak ) )
p90RSK = ( Erk AND ( ( ( PDK1 ) ) AND ( ( NOT p90RSK ) ) ) )
EGFR_EGFR_EGF_MVB = ( ( EGFR_EGFR_EGF_MVB ) AND NOT ( EGFR_EGFR_EGF_Lysosome ) ) OR ( EGFR_EGFR_EGF_End )
Spry2 = ( ( EGFR_ErbB3 ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_ErbB2 ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR_TGFa_CCV ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR_EGF_End ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_ErbB4 ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR_EGF_CCP ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR_EGF_CCV ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR_TGFa_CCP ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR_EGF_MVB ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR_EGF_PM ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR_TGFa_End ) AND NOT ( Cbl_RTK ) ) OR ( ( EGFR_EGFR_TGFa_PM ) AND NOT ( Cbl_RTK ) )
ESCRT_I = ( ESCRT_0 )
VPS4 = ( ESCRT_III )
UBPY = ( Alix ) OR ( ESCRT_III ) OR ( ESCRT_I )
MLK1 = ( Cdc42 ) OR ( Rac )
cAMP = ( ( cAMP ) AND NOT ( PDE4 ) ) OR ( ( AC ) AND NOT ( PDE4 ) )
PLD = ( Rho AND ( ( ( Actin ) AND ( ( ( PIP3_345 ) ) OR ( ( PIP2_45 ) ) ) ) AND ( ( NOT ARF ) ) ) ) OR ( PKC AND ( ( ( Actin ) AND ( ( ( PIP3_345 ) ) OR ( ( PIP2_45 ) ) ) ) AND ( ( NOT ARF ) ) ) ) OR ( ARF AND ( ( ( PIP2_45 ) ) OR ( ( PIP3_345 ) ) ) ) OR ( Rac AND ( ( ( NOT ARF ) ) AND ( ( Actin ) AND ( ( ( PIP2_45 ) ) OR ( ( PIP3_345 ) ) ) ) ) ) OR ( Cdc42 AND ( ( ( NOT ARF ) ) AND ( ( Actin ) AND ( ( ( PIP2_45 ) ) OR ( ( PIP3_345 ) ) ) ) ) )
Fak = ( ( Integrins AND ( ( ( Talin ) ) ) ) AND NOT ( PTEN AND ( ( ( Fak ) ) ) ) ) OR ( ( Src AND ( ( ( Fak ) ) ) ) AND NOT ( PTEN AND ( ( ( Fak ) ) ) ) )
Nck = ( EGFR_ErbB3 ) OR ( EGFR_EGFR_TGFa_CCV ) OR ( EGFR_ErbB2 ) OR ( EGFR_EGFR_EGF_End ) OR ( EGFR_ErbB4 ) OR ( Cas ) OR ( EGFR_EGFR_EGF_CCP ) OR ( EGFR_EGFR_EGF_CCV ) OR ( EGFR_EGFR_TGFa_CCP ) OR ( EGFR_EGFR_EGF_MVB ) OR ( EGFR_EGFR ) OR ( EGFR_EGFR_TGFa_End ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
ErbB2_Ub = ( Cbl_ErbB2 AND ( ( ( NOT Hsp90 ) ) ) ) OR ( CHIP )
EGFR_Y1101 = ( Src )
EGFR_ErbB3 = ( EGF AND ( ( ( NRG ) ) AND ( ( NOT ErbB2_Free ) ) AND ( ( NOT EGFR_T654 ) ) AND ( ( EGFR_Free ) ) AND ( ( ErbB3_Free ) ) ) )
PTPPEST = ( ( ( Integrins AND ( ( ( ECM ) ) ) ) AND NOT ( PKA ) ) AND NOT ( PKC ) )
Ral = ( CaM ) OR ( AND_34 ) OR ( RalGDS )
Gai = ( Gbg_i AND ( ( ( NOT RGS ) ) AND ( ( Gai ) ) ) ) OR ( alpha_iR AND ( ( ( NOT Gai AND NOT Gbg_i ) ) ) ) OR ( PKA AND ( ( ( NOT Gbg_i ) ) AND ( ( NOT Gai ) ) AND ( ( alpha_sL ) ) AND ( ( NOT alpha_sR ) ) ) )
PAK = ( ( Rac AND ( ( ( Grb2 ) ) OR ( ( Nck ) AND ( ( ( NOT Akt ) ) ) ) ) ) AND NOT ( PKA ) ) OR ( ( ( Src AND ( ( ( PAK ) AND ( ( ( Cdc42 OR Rac ) ) ) ) ) ) AND NOT ( PTP1b ) ) AND NOT ( PKA ) ) OR ( ( Cdc42 AND ( ( ( Grb2 ) ) OR ( ( Nck ) AND ( ( ( NOT Akt ) ) ) ) ) ) AND NOT ( PKA ) )
EGFR_T669 = ( Erk )
ErbB3_Free = ( ErbB3_Contr ) OR ( ( ( ( ErbB3_Free ) AND NOT ( EGFR_ErbB3 ) ) AND NOT ( ErbB2_ErbB3 ) ) AND NOT ( ErbB3_ErbB4 ) )
NIK = ( TAK1 ) OR ( Nck )
MLCK = ( ( ( CaM AND ( ( ( NOT PKA ) ) AND ( ( NOT PAK ) ) ) ) AND NOT ( PAK ) ) AND NOT ( PKA ) ) OR ( ( ( Erk AND ( ( ( NOT PKA ) ) AND ( ( NOT PAK ) ) ) ) AND NOT ( PAK ) ) AND NOT ( PKA ) )
PLC_g = ( AA ) OR ( EGFR_Y1068 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR EGFR_EGFR_EGF_End ) ) ) ) OR ( EGFR_Y992 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR EGFR_EGFR_EGF_End ) ) ) ) OR ( EGFR_Y1173 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR EGFR_EGFR_EGF_End ) ) ) )
Ras = ( RasGRF_GRP ) OR ( SHP2 ) OR ( Sos )
p115RhoGEF = ( Ga_1213 AND ( ( ( PIP3_345 ) ) ) )
Rho = ( Rho AND ( ( ( NOT p190RhoGAP AND NOT Graf AND NOT PKA ) ) ) ) OR ( p115RhoGEF AND ( ( ( NOT Rho AND NOT RhoGDI ) ) AND ( ( p120_catenin ) ) ) )
Integrins = ( Src AND ( ( ( NOT Integrins AND NOT Talin AND NOT ECM AND NOT ILK AND NOT PP2A ) ) ) ) OR ( Talin AND ( ( ( ECM ) ) AND ( ( NOT Integrins AND NOT ILK ) ) ) ) OR ( PP2A AND ( ( ( Talin AND ECM AND ILK ) ) AND ( ( NOT Integrins ) ) ) ) OR ( Integrins AND ( ( ( NOT ILK AND NOT Src ) ) ) )
GRK = ( ( ( Gbg_q AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) ) OR ( ( ( Gbg_i AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) ) OR ( ( ( B_Arrestin AND ( ( ( Src ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) ) OR ( ( ( Gbg_1213 AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) ) OR ( ( ( Gbg_s AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( Erk ) ) AND NOT ( RKIP ) )
PIP3_345 = ( ( PI5K AND ( ( ( PIP2_34 ) ) ) ) AND NOT ( PTEN AND ( ( ( PIP3_345 ) ) ) ) ) OR ( ( PI3K AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( PTEN AND ( ( ( PIP3_345 ) ) ) ) )
EGFR_EGFR_TGFa_PM = ( ( ( EGFR_Free AND ( ( ( NOT EGFR_T654 ) ) AND ( ( TGFa ) ) ) ) AND NOT ( ErbB2_Free ) ) AND NOT ( EGFR_EGFR_TGFa_CCP ) ) OR ( ( EGFR_EGFR_TGFa_PM ) AND NOT ( EGFR_EGFR_TGFa_CCP ) )
MKK7 = ( Mekk4 AND ( ( ( ASK1 ) ) ) ) OR ( MLK1 AND ( ( ( ASK1 ) ) ) ) OR ( MLK2 AND ( ( ( ASK1 ) ) ) ) OR ( MLK3 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk1 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk3 AND ( ( ( ASK1 ) ) ) )
ErbB2_Y1248 = ( EGFR_ErbB2 ) OR ( ErbB2_ErbB3 ) OR ( ErbB2_ErbB4 )
WASP = ( ( Src AND ( ( ( PIP2_45 OR Nck OR Grb2 ) ) AND ( ( Cdc42 AND Crk ) ) ) ) AND NOT ( PTPPEST ) ) OR ( ( Fak AND ( ( ( Cdc42 AND Crk ) ) AND ( ( PIP2_45 OR Nck OR Grb2 ) ) ) ) AND NOT ( PTPPEST ) ) OR ( ( Cdc42 AND ( ( ( Src OR Fak ) ) AND ( ( NOT PTPPEST AND NOT Crk ) ) AND ( ( PIP2_45 OR Nck OR Grb2 ) ) ) ) AND NOT ( PTPPEST ) )
PTPa = ( PKC )
IQGAP1 = ( NOT ( ( CaM AND ( ( ( Ca ) ) ) ) ) ) OR NOT ( Ca OR CaM )
Eps15 = ( EGFR_Ub ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
ErbB2_Y1196 = ( EGFR_ErbB2 ) OR ( ErbB2_ErbB3 ) OR ( ErbB2_ErbB4 )
IP3R1 = ( ( ( ( Gbg_i ) AND NOT ( Ca AND ( ( ( IP3R1 ) ) AND ( ( NOT IP3 ) ) ) ) ) AND NOT ( CaM AND ( ( ( Ca ) ) AND ( ( IP3R1 ) ) ) ) ) AND NOT ( IP3R1 AND ( ( ( NOT Ca AND NOT IP3 AND NOT PKA AND NOT PP2A ) ) AND ( ( CaM AND Gbg_i ) ) ) ) ) OR ( ( ( IP3 AND ( ( ( Ca ) ) ) ) AND NOT ( Ca AND ( ( ( IP3R1 ) ) AND ( ( NOT IP3 ) ) ) ) ) AND NOT ( CaM AND ( ( ( Ca ) ) AND ( ( IP3R1 ) ) ) ) ) OR ( ( ( ( PKA ) AND NOT ( Ca AND ( ( ( IP3R1 ) ) AND ( ( NOT IP3 ) ) ) ) ) AND NOT ( PP2A AND ( ( ( IP3R1 ) ) ) ) ) AND NOT ( CaM AND ( ( ( Ca ) ) AND ( ( IP3R1 ) ) ) ) )
GCK = ( Trafs )
TAK1 = ( Tab_12 )
EGFR_EGFR_TGFa_CCV = ( ( EGFR_EGFR_TGFa_CCV ) AND NOT ( EGFR_EGFR_TGFa_End ) ) OR ( EGFR_EGFR_TGFa_CCP AND ( ( ( GAK OR Clathrin ) ) AND ( ( Dynamin AND Actin ) ) AND ( ( PIP2_45 AND AP2 ) ) ) )
EGFR_EGFR_EGF_CCV = ( ( EGFR_EGFR_EGF_CCV ) AND NOT ( EGFR_EGFR_EGF_End ) ) OR ( ( EGFR_EGFR_EGF_CCP AND ( ( ( Dynamin AND Actin ) ) ) ) AND NOT ( EGFR_EGFR_EGF_End ) )
Rab5 = ( ( p120RasGAP ) AND NOT ( Rab7 AND ( ( ( Rab5 ) ) ) ) ) OR ( ( Rab5 ) AND NOT ( Rab7 AND ( ( ( Rab5 ) ) ) ) ) OR ( ( Rabex_5 ) AND NOT ( Rab7 AND ( ( ( Rab5 ) ) ) ) ) OR ( ( EGFR_EGFR_EGF_PM ) AND NOT ( Rab7 AND ( ( ( Rab5 ) ) ) ) ) OR ( ( RIN ) AND NOT ( Rab7 AND ( ( ( Rab5 ) ) ) ) ) OR ( ( EGFR_EGFR_TGFa_PM ) AND NOT ( Rab7 AND ( ( ( Rab5 ) ) ) ) )
CHIP = ( AG AND ( ( ( Hsp90 ) ) ) )
DOCK180 = ( Crk AND ( ( ( Cas ) ) AND ( ( PIP3_345 ) ) ) )
RhoGDI = ( NOT ( ( AA ) OR ( PKC ) OR ( PIP2_45 ) ) ) OR NOT ( AA OR PIP2_45 OR PKC )
Raf_DeP = ( PP2A AND ( ( ( Raf_Rest ) ) AND ( ( NOT Raf_DeP ) ) ) ) OR ( Raf_DeP AND ( ( ( NOT Raf_Loc ) ) ) )
Clathrin = ( GAK ) OR ( Hip1R ) OR ( CALM AND ( ( ( PIP2_45 ) ) ) ) OR ( AP2 ) OR ( Epsin AND ( ( ( PIP2_45 ) ) ) ) OR ( Src ) OR ( ESCRT_0 )
Graf = ( Fak AND ( ( ( Src ) ) ) )
ErbB2_ErbB4 = ( NRG AND ( ( ( ErbB4_Free AND ErbB2_Free ) AND ( ( ( NOT ErbB3_Free ) ) ) ) ) )
Crk = ( ( Cas AND ( ( ( Src OR Fak ) ) ) ) AND NOT ( PTPPEST ) )
Trx = ( Stress ) OR ( Trafs )
Sek1 = ( Mekk4 AND ( ( ( ASK1 ) ) ) ) OR ( MLK1 AND ( ( ( ASK1 ) ) ) ) OR ( MLK2 AND ( ( ( ASK1 ) ) ) ) OR ( MLK3 AND ( ( ( ASK1 ) ) ) ) OR ( TAK1 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk1 AND ( ( ( ASK1 ) ) ) ) OR ( Tpl2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk3 AND ( ( ( ASK1 ) ) ) )
Palpha_1213R = ( alpha_1213R AND ( ( ( GRK ) ) ) )
PDK1 = ( p90RSK ) OR ( Src )
PI4K = ( Rho ) OR ( PKC ) OR ( ARF ) OR ( Gai ) OR ( Gaq )
EGFR_Y891 = ( Src )
MLK3 = ( IL1_TNFR ) OR ( Rac ) OR ( Cdc42 )
PKA = ( ( PKA AND ( ( ( cAMP ) ) ) ) AND NOT ( PP2A AND ( ( ( PKA ) ) ) ) ) OR ( ( PDK1 AND ( ( ( cAMP ) ) ) ) AND NOT ( PP2A AND ( ( ( PKA ) ) ) ) )
Rab7 = ( Rab5 )
Rac = ( ( RasGRF_GRP AND ( ( ( Integrins AND ECM ) ) ) ) AND NOT ( RalBP1 AND ( ( ( Rac ) ) ) ) ) OR ( ( Rac AND ( ( ( NOT RalBP1 ) ) ) ) AND NOT ( RalBP1 AND ( ( ( Rac ) ) ) ) ) OR ( Pix_Cool AND ( ( ( NOT PAK ) AND ( ( ( Cdc42 ) ) AND ( ( Integrins AND ECM ) ) AND ( ( NOT Tiam AND NOT Rac AND NOT RasGRF_GRP AND NOT DOCK180 ) ) ) ) OR ( ( PAK AND Gbg_i ) AND ( ( ( Integrins AND ECM ) ) AND ( ( NOT Rac ) ) ) ) OR ( ( NOT Gbg_i ) AND ( ( ( Cdc42 ) ) AND ( ( NOT Rac ) ) AND ( ( Integrins AND ECM ) ) ) ) ) ) OR ( ( Tiam AND ( ( ( Integrins AND ECM ) ) ) ) AND NOT ( RalBP1 AND ( ( ( Rac ) ) ) ) ) OR ( ( DOCK180 AND ( ( ( Integrins AND ECM ) ) ) ) AND NOT ( RalBP1 AND ( ( ( Rac ) ) ) ) )
ESCRT_0 = ( EGFR_EGFR_EGF_End AND ( ( ( PIP3_345 ) ) ) )
Gbg_i = ( Gai ) OR ( alpha_iR AND ( ( ( NOT Gbg_i ) ) AND ( ( NOT Gai ) ) ) )
CaMK = ( CaMKK AND ( ( ( CaM ) ) ) )
ErbB4_Y1188 = ( ErbB4_ErbB4 ) OR ( ErbB3_ErbB4 ) OR ( ErbB2_ErbB4 ) OR ( EGFR_ErbB4 )
Raf_Loc = ( Raf_Loc AND ( ( ( NOT Raf ) ) ) ) OR ( Ras AND ( ( ( NOT Raf_Loc ) ) AND ( ( Raf_DeP ) ) ) )
Gbg_q = ( alpha_qR AND ( ( ( NOT Gbg_q ) ) AND ( ( NOT Gaq ) ) ) ) OR ( Gaq )
Shc = ( ( EGFR_Y992 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR ErbB3_ErbB4 OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR ErbB2_ErbB4 OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( PTEN ) ) OR ( ( Fak ) AND NOT ( PTEN ) ) OR ( ( EGFR_Y1173 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR ErbB3_ErbB4 OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR ErbB2_ErbB4 OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( PTEN ) ) OR ( ( ErbB2_Y1196 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR ErbB3_ErbB4 OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR ErbB2_ErbB4 OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( PTEN ) ) OR ( ( Src ) AND NOT ( PTEN ) ) OR ( ( ErbB4_Y1242 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR ErbB3_ErbB4 OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR ErbB2_ErbB4 OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( PTEN ) ) OR ( ( ErbB4_Y1188 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR ErbB3_ErbB4 OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR ErbB2_ErbB4 OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( PTEN ) ) OR ( ( ErbB2_Y1221_22 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR ErbB3_ErbB4 OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR ErbB2_ErbB4 OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( PTEN ) ) OR ( ( ErbB3_Y1309 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR ErbB3_ErbB4 OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR ErbB2_ErbB4 OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( PTEN ) ) OR ( ( ErbB2_Y1248 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR ErbB3_ErbB4 OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR ErbB2_ErbB4 OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( PTEN ) ) OR ( ( EGFR_Y1148 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR ErbB3_ErbB4 OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR ErbB2_ErbB4 OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( PTEN ) )
DGK = ( EGFR_ErbB3 ) OR ( EGFR_ErbB2 ) OR ( EGFR_EGFR_TGFa_CCV ) OR ( EGFR_EGFR_EGF_End ) OR ( EGFR_ErbB4 ) OR ( Src AND ( ( ( Ca AND PA ) ) ) ) OR ( EGFR_EGFR_EGF_CCP ) OR ( EGFR_EGFR_EGF_CCV ) OR ( PKC AND ( ( ( DAG ) ) ) ) OR ( EGFR_EGFR_TGFa_CCP ) OR ( EGFR_EGFR_EGF_MVB ) OR ( EGFR_EGFR ) OR ( EGFR_EGFR_TGFa_End ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
AND_34 = ( Cas )
Rabaptin_5 = ( Rab5 )
RalBP1 = ( Ral )
Cbl_ErbB2 = ( Trastuzumab AND ( ( ( ErbB2_ErbB4 ) ) OR ( ( ErbB2_ErbB3 ) ) OR ( ( EGFR_ErbB2 ) ) ) )
Actin = ( Arp_23 AND ( ( ( alpha_catenin ) ) AND ( ( NOT IQGAP1 ) ) AND ( ( Myosin ) ) ) ) OR ( IQGAP1 AND ( ( ( Myosin ) ) ) )
MKPs = ( p38 AND ( ( ( cAMP ) ) ) ) OR ( Erk AND ( ( ( cAMP ) ) ) ) OR ( SAPK AND ( ( ( cAMP ) ) ) )
Fer = ( E_cadherin AND ( ( ( p120_catenin ) ) ) )
Rap1 = ( CaMK AND ( ( ( NOT Gai OR NOT Rap1 ) ) AND ( ( cAMP AND Src ) ) ) ) OR ( PKA AND ( ( ( NOT Gai OR NOT Rap1 ) ) AND ( ( cAMP AND Src ) ) ) )
RasGRF_GRP = ( CaM AND ( ( ( Cdc42 ) ) ) ) OR ( DAG AND ( ( ( Cdc42 ) ) ) )
ErbB2_Y1139 = ( EGFR_ErbB2 ) OR ( ErbB2_ErbB3 ) OR ( ErbB2_ErbB4 )
IP3 = ( PLC_B AND ( ( ( PIP2_45 ) ) ) ) OR ( PLC_g AND ( ( ( PIP2_45 ) ) ) )
Dynamin = ( Grb2 ) OR ( Endophilin ) OR ( EGFR_EGFR_EGF_PM ) OR ( PIP2_45 ) OR ( EGFR_EGFR_TGFa_PM )
Akt = ( CaMKK AND ( ( ( PIP3_345 OR PIP2_34 ) ) AND ( ( ILK AND Src ) ) AND ( ( NOT Akt ) ) ) ) OR ( Akt AND ( ( ( NOT PP2A ) ) ) ) OR ( PDK1 AND ( ( ( ILK AND Src ) ) AND ( ( NOT Akt ) ) AND ( ( PIP3_345 OR PIP2_34 ) ) ) )
Tpl2 = ( Trafs )
E_cadherin = ( ( ( B_catenin AND ( ( ( ExtE_cadherin ) ) ) ) AND NOT ( IQGAP1 AND ( ( ( NOT Cdc42 AND NOT Rac ) ) ) ) ) AND NOT ( Hakai AND ( ( ( NOT p120_catenin ) ) ) ) )
p38 = ( ( ( MKK6 ) AND NOT ( PP2A ) ) AND NOT ( MKPs ) ) OR ( ( ( MKK3 ) AND NOT ( PP2A ) ) AND NOT ( MKPs ) ) OR ( ( ( Sek1 ) AND NOT ( PP2A ) ) AND NOT ( MKPs ) )
Raf_Rest = ( ( Raf_Rest AND ( ( ( NOT Raf_DeP ) ) ) ) OR ( Raf_DeP AND ( ( ( NOT Raf_Rest AND NOT Raf ) ) ) ) ) OR NOT ( Raf_DeP OR Raf_Rest OR Raf )
EGFR_Ub = ( ( ( EGFR_EGFR_EGF_PM AND ( ( ( Cbl_RTK ) ) ) ) AND NOT ( EGFR_EGFR_EGF_CCP ) ) AND NOT ( EGFR_EGFR_TGFa_CCP ) ) OR ( ( ( EGFR_EGFR_TGFa_PM AND ( ( ( Cbl_RTK ) ) ) ) AND NOT ( EGFR_EGFR_EGF_CCP ) ) AND NOT ( EGFR_EGFR_TGFa_CCP ) )
Cbp = ( ( Src ) AND NOT ( SHP2 ) )
MKK6 = ( Mekk4 AND ( ( ( ASK1 ) ) ) ) OR ( MLK3 AND ( ( ( ASK1 ) ) ) ) OR ( PAK AND ( ( ( ASK1 ) ) ) ) OR ( TAK1 AND ( ( ( ASK1 ) ) ) ) OR ( Tpl2 AND ( ( ( ASK1 ) ) ) ) OR ( TAO_12 AND ( ( ( ASK1 ) ) ) )
Rabex_5 = ( Rabaptin_5 )
TAO_12 = ( Stress )
alpha_qR = ( ( alpha_qL ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_iR AND NOT alpha_qL AND NOT alpha_qR ) ) OR ( ( Palpha_iR ) ) ) ) ) OR ( ( Palpha_iR AND ( ( ( NOT B_Arrestin ) ) ) ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_iR AND NOT alpha_qL AND NOT alpha_qR ) ) OR ( ( Palpha_iR ) ) ) ) ) OR ( ( alpha_qR ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_iR AND NOT alpha_qL AND NOT alpha_qR ) ) OR ( ( Palpha_iR ) ) ) ) )
p120_catenin = ( EGFR_ErbB3 ) OR ( EGFR_EGFR_TGFa_CCV ) OR ( EGFR_ErbB2 ) OR ( EGFR_EGFR_EGF_End ) OR ( EGFR_ErbB4 ) OR ( Src ) OR ( EGFR_EGFR_EGF_CCP ) OR ( ( Rho ) AND NOT ( Fer ) ) OR ( EGFR_EGFR_EGF_CCV ) OR ( EGFR_EGFR_TGFa_CCP ) OR ( EGFR_EGFR_EGF_MVB ) OR ( EGFR_EGFR ) OR ( EGFR_EGFR_TGFa_End ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
Arp_23 = ( WASP )
Cas = ( ( Src AND ( ( ( Fak ) ) ) ) AND NOT ( PTPPEST AND ( ( ( Cas ) ) ) ) )
p190RhoGAP = ( Src AND ( ( ( NOT p190RhoGAP ) ) OR ( ( NOT p120RasGAP ) ) OR ( ( Fak ) ) ) ) OR ( Fak AND ( ( ( Src ) ) ) )
Csk = ( Cbp AND ( ( ( Gbg_1213 OR PKA OR Gbg_q OR Gbg_i ) ) OR ( ( NOT SHP2 AND NOT Gbg_1213 AND NOT PKA AND NOT Gbg_q AND NOT Gbg_i ) ) ) ) OR ( ( Fak AND ( ( ( Cbp AND Src ) ) ) ) AND NOT ( SHP2 ) )
ESCRT_II = ( ESCRT_I )
CIN85 = ( Cbl_RTK )
Raf = ( Ras AND ( ( ( Raf ) ) ) ) OR ( Src AND ( ( ( NOT Raf ) ) AND ( ( PAK AND Raf_Loc AND RKIP ) ) ) ) OR ( Raf AND ( ( ( NOT Akt AND NOT PKA AND NOT Erk ) ) ) ) OR ( PAK AND ( ( ( Raf ) ) AND ( ( NOT Ras AND NOT Akt AND NOT Erk ) ) ) )
ErbB2_Y1023 = ( EGFR_ErbB2 ) OR ( ErbB2_ErbB3 ) OR ( ErbB2_ErbB4 )
Palpha_iR = ( alpha_iR AND ( ( ( GRK ) ) ) )
IL1_TNFR = ( IL1_TNF )
alpha_1213R = ( ( alpha_1213L ) AND NOT ( B_Arrestin AND ( ( ( NOT alpha_1213R AND NOT alpha_1213L AND NOT Palpha_1213R ) ) OR ( ( Palpha_1213R ) ) ) ) ) OR ( ( Palpha_1213R AND ( ( ( NOT B_Arrestin ) ) ) ) AND NOT ( B_Arrestin AND ( ( ( NOT alpha_1213R AND NOT alpha_1213L AND NOT Palpha_1213R ) ) OR ( ( Palpha_1213R ) ) ) ) ) OR ( ( alpha_1213R ) AND NOT ( B_Arrestin AND ( ( ( NOT alpha_1213R AND NOT alpha_1213L AND NOT Palpha_1213R ) ) OR ( ( Palpha_1213R ) ) ) ) )
MKK3 = ( Mekk4 AND ( ( ( ASK1 ) ) ) ) OR ( MLK1 AND ( ( ( ASK1 ) ) ) ) OR ( MLK2 AND ( ( ( ASK1 ) ) ) ) OR ( MLK3 AND ( ( ( ASK1 ) ) ) ) OR ( TAK1 AND ( ( ( ASK1 ) ) ) ) OR ( Tpl2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk2 AND ( ( ( ASK1 ) ) ) ) OR ( Mekk3 AND ( ( ( ASK1 ) ) ) ) OR ( PAK AND ( ( ( ASK1 ) ) ) ) OR ( TAO_12 AND ( ( ( ASK1 ) ) ) )
PTP1b = ( NOT ( ( EGFR_ErbB3 ) OR ( EGFR_ErbB2 ) OR ( EGFR_EGFR_TGFa_CCV ) OR ( EGFR_EGFR_EGF_End ) OR ( EGFR_ErbB4 ) OR ( EGFR_EGFR_EGF_CCP ) OR ( EGFR_EGFR_EGF_CCV ) OR ( EGFR_EGFR_TGFa_CCP ) OR ( EGFR_EGFR_EGF_MVB ) OR ( EGFR_EGFR ) OR ( EGFR_EGFR_EGF_PM ) OR ( Stress ) OR ( EGFR_EGFR_TGFa_PM ) ) ) OR NOT ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR Stress OR EGFR_ErbB4 OR EGFR_EGFR OR EGFR_EGFR_EGF_End )
AMSH = ( Alix ) OR ( ESCRT_0 ) OR ( ESCRT_I )
Trafs = ( IL1_TNFR )
AC = ( Integrins AND ( ( ( ECM ) AND ( ( ( Gas ) ) AND ( ( Gbg_i ) ) ) ) ) )
EGFR_EGFR_EGF_CCP = ( ( Eps15 AND ( ( ( EGFR_EGFR_EGF_PM ) ) AND ( ( Rab5 ) ) AND ( ( PIP2_45 ) ) AND ( ( Cbl_RTK ) ) AND ( ( Clathrin ) ) ) ) AND NOT ( EGFR_EGFR_EGF_CCV ) ) OR ( ( Epsin AND ( ( ( Cbl_RTK ) ) AND ( ( EGFR_EGFR_EGF_PM ) ) AND ( ( Rab5 ) ) AND ( ( PIP2_45 ) ) AND ( ( Clathrin ) ) ) ) AND NOT ( EGFR_EGFR_EGF_CCV ) ) OR ( ( AP2 AND ( ( ( Rab5 ) ) AND ( ( Clathrin ) ) AND ( ( EGFR_EGFR_EGF_PM ) ) AND ( ( Cbl_RTK ) ) AND ( ( PIP2_45 ) ) ) ) AND NOT ( EGFR_EGFR_EGF_CCV ) ) OR ( ( EGFR_EGFR_EGF_CCP ) AND NOT ( EGFR_EGFR_EGF_CCV ) )
EGFR_Y1086 = ( EGFR_ErbB3 ) OR ( EGFR_ErbB2 ) OR ( EGFR_ErbB4 ) OR ( EGFR_EGFR ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
EGFR_EGFR = ( alpha_qR AND ( ( ( EGFR_Free ) ) AND ( ( Ca ) ) AND ( ( EGFR_T654 ) ) ) ) OR ( alpha_iR AND ( ( ( Ca ) ) AND ( ( EGFR_Free ) ) AND ( ( EGFR_T654 ) ) ) ) OR ( alpha_1213R AND ( ( ( Ca ) ) AND ( ( EGFR_Free ) ) AND ( ( EGFR_T654 ) ) ) )
Gab1 = ( ( Gab1 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR EGFR_EGFR_EGF_End ) ) AND ( ( PIP3_345 ) ) ) ) AND NOT ( SHP2 ) ) OR ( ( Grb2 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR EGFR_EGFR_EGF_End ) ) AND ( ( NOT Gab1 ) ) ) ) AND NOT ( SHP2 ) )
Palpha_sR = ( alpha_sR AND ( ( ( GRK ) ) ) )
EEA1 = ( Rab5 AND ( ( ( PIP3_345 ) ) ) )
CALM = ( PIP2_45 )
PTEN = ( ( Stress ) AND NOT ( PTEN_I ) ) OR ( ( Pix_Cool AND ( ( ( Rho ) ) AND ( ( Cdc42 ) ) AND ( ( PI3K ) ) ) ) AND NOT ( PTEN_I ) )
B_Arrestin = ( Palpha_iR ) OR ( Palpha_qR ) OR ( Palpha_1213R ) OR ( Palpha_sR )
ErbB4_Y1056 = ( ErbB4_ErbB4 ) OR ( ErbB3_ErbB4 ) OR ( EGFR_ErbB4 ) OR ( ErbB2_ErbB4 )
SHP2 = ( Gab1 )
ErbB3_Y1257 = ( EGFR_ErbB3 ) OR ( ErbB3_ErbB4 ) OR ( ErbB2_ErbB3 )
Palpha_qR = ( alpha_qR AND ( ( ( GRK ) ) ) )
ErbB4_ErbB4 = ( ( NRG AND ( ( ( ErbB4_Free ) ) ) ) AND NOT ( ErbB2_Free ) )
EGFR_Y1045 = ( EGFR_ErbB3 ) OR ( EGFR_ErbB2 ) OR ( EGFR_ErbB4 ) OR ( EGFR_EGFR ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
EGFR_Y845 = ( EGFR_Free AND ( ( ( Cas AND Integrins AND Src ) ) ) ) OR ( Src AND ( ( ( EGFR_EGFR_EGF_PM ) ) ) )
Gbg_s = ( Gas ) OR ( alpha_sR AND ( ( ( NOT Gbg_s ) ) AND ( ( NOT Gas ) ) ) )
ErbB3_Y1243 = ( EGFR_ErbB3 ) OR ( ErbB3_ErbB4 ) OR ( ErbB2_ErbB3 )
EGFR_Y1068 = ( EGFR_ErbB3 ) OR ( EGFR_ErbB2 ) OR ( EGFR_ErbB4 ) OR ( EGFR_EGFR ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
ESCRT_III = ( ESCRT_II ) OR ( ESCRT_I )
AA = ( PLA2 )
PLA2 = ( PIP3_345 AND ( ( ( PIP2_45 ) ) AND ( ( CaMK ) ) ) ) OR ( PIP2_45 AND ( ( ( Erk ) ) AND ( ( PIP3_345 ) ) ) ) OR ( Erk AND ( ( ( Ca ) ) ) ) OR ( CaMK AND ( ( ( Ca ) ) ) )
EGFR_EGFR_EGF_SR = ( EGFR_EGFR_EGF_MVB AND ( ( ( AMSH ) ) ) )
Cbl_FA = ( ( Src AND ( ( ( Pix_Cool ) ) AND ( ( Cdc42 ) ) ) ) AND NOT ( Cbl_RTK ) )
EGFR_Free = ( ( EGFR_EGFR_TGFa_End ) AND NOT ( EGFR_Free AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_PM OR EGFR_ErbB3 OR EGFR_EGFR OR EGFR_EGFR_EGF_PM ) ) ) ) ) OR ( ( EGFR_Free ) AND NOT ( EGFR_Free AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_PM OR EGFR_ErbB3 OR EGFR_EGFR OR EGFR_EGFR_EGF_PM ) ) ) ) ) OR ( ( EGFR_Contr ) AND NOT ( EGFR_Free AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_PM OR EGFR_ErbB3 OR EGFR_EGFR OR EGFR_EGFR_EGF_PM ) ) ) ) )
alpha_iR = ( ( alpha_iL ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_iR AND NOT alpha_iL AND NOT alpha_iR ) ) OR ( ( Palpha_iR ) ) ) ) ) OR ( ( Palpha_iR AND ( ( ( NOT B_Arrestin ) ) ) ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_iR AND NOT alpha_iL AND NOT alpha_iR ) ) OR ( ( Palpha_iR ) ) ) ) ) OR ( ( alpha_iR ) AND NOT ( B_Arrestin AND ( ( ( NOT Palpha_iR AND NOT alpha_iL AND NOT alpha_iR ) ) OR ( ( Palpha_iR ) ) ) ) )
Vinc = ( Actin AND ( ( ( Talin AND Vinc ) ) AND ( ( NOT PIP2_45 ) ) ) ) OR ( Talin AND ( ( ( Src ) ) ) )
Ca = ( ( IP3R1 ) AND NOT ( ExtPump ) )
ErbB3_Y1203_05 = ( EGFR_ErbB3 ) OR ( ErbB3_ErbB4 ) OR ( ErbB2_ErbB3 )
Erk = ( Mek ) OR ( ( ( Erk ) AND NOT ( MKPs ) ) AND NOT ( PP2A ) )
PP2A = ( ( ( EGFR_ErbB3 ) AND NOT ( PP2A ) ) OR ( ( EGFR_ErbB2 ) AND NOT ( PP2A ) ) OR ( ( EGFR_EGFR_TGFa_CCV ) AND NOT ( PP2A ) ) OR ( ( EGFR_EGFR_EGF_End ) AND NOT ( PP2A ) ) OR ( ( EGFR_ErbB4 ) AND NOT ( PP2A ) ) OR ( ( EGFR_EGFR_EGF_CCP ) AND NOT ( PP2A ) ) OR ( ( EGFR_EGFR_EGF_CCV ) AND NOT ( PP2A ) ) OR ( PP2A AND ( ( ( NOT EGFR_ErbB2 AND NOT EGFR_EGFR_TGFa_CCV AND NOT EGFR_EGFR_TGFa_PM AND NOT EGFR_EGFR_EGF_CCV AND NOT EGFR_ErbB3 AND NOT EGFR_EGFR_TGFa_End AND NOT EGFR_EGFR_EGF_PM AND NOT EGFR_EGFR_EGF_MVB AND NOT EGFR_EGFR_TGFa_CCP AND NOT EGFR_EGFR_EGF_CCP AND NOT EGFR_ErbB4 AND NOT EGFR_EGFR AND NOT EGFR_EGFR_EGF_End ) ) ) ) OR ( ( EGFR_EGFR_TGFa_CCP ) AND NOT ( PP2A ) ) OR ( ( EGFR_EGFR_EGF_MVB ) AND NOT ( PP2A ) ) OR ( ( EGFR_EGFR ) AND NOT ( PP2A ) ) OR ( ( EGFR_EGFR_EGF_PM ) AND NOT ( PP2A ) ) OR ( ( EGFR_EGFR_TGFa_End ) AND NOT ( PP2A ) ) OR ( ( EGFR_EGFR_TGFa_PM ) AND NOT ( PP2A ) ) ) OR NOT ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_TGFa_End OR PP2A OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_EGFR OR EGFR_EGFR_EGF_End )
ErbB2_Free = ( ( ErbB2_Contr ) AND NOT ( Trastuzumab ) ) OR ( ( ( ( ( ErbB2_Free ) AND NOT ( Trastuzumab ) ) AND NOT ( EGFR_ErbB2 ) ) AND NOT ( ErbB2_ErbB3 ) ) AND NOT ( ErbB2_ErbB4 ) )
PI5K = ( PA ) OR ( PI5K AND ( ( ( Talin ) ) ) ) OR ( RhoK ) OR ( ARF ) OR ( Src AND ( ( ( NOT Talin ) ) AND ( ( NOT PI5K ) ) AND ( ( Fak ) ) ) )
PIP_4 = ( ( ( PTEN AND ( ( ( NOT PIP_4 ) ) AND ( ( PIP2_34 ) ) ) ) AND NOT ( PI5K AND ( ( ( PIP_4 ) ) ) ) ) AND NOT ( PI3K AND ( ( ( PIP_4 ) ) ) ) ) OR ( ( ( PIP_4 AND ( ( ( NOT PI5K ) ) AND ( ( NOT PI3K ) ) ) ) AND NOT ( PI5K AND ( ( ( PIP_4 ) ) ) ) ) AND NOT ( PI3K AND ( ( ( PIP_4 ) ) ) ) ) OR ( ( ( PI4K AND ( ( ( NOT PIP_4 ) ) ) ) AND NOT ( PI5K AND ( ( ( PIP_4 ) ) ) ) ) AND NOT ( PI3K AND ( ( ( PIP_4 ) ) ) ) )
PKC_primed = ( PKC_primed AND ( ( ( NOT PKC ) ) ) ) OR ( PKC AND ( ( ( NOT PKC_primed ) ) AND ( ( PDK1 ) ) ) ) OR ( PDK1 AND ( ( ( NOT PKC ) ) ) )
ErbB2_Lysosome = ( ErbB2_ErbB3 AND ( ( ( ErbB2Deg_Contr ) ) ) ) OR ( ErbB2_Ub AND ( ( ( NOT ErbB2Deg_Contr ) ) ) )
ErbB3_Y1309 = ( EGFR_ErbB3 ) OR ( ErbB3_ErbB4 ) OR ( ErbB2_ErbB3 )
CaM = ( Ca )
Pix_Cool = ( PIP3_345 AND ( ( ( B_Parvin ) ) ) ) OR ( PIP2_34 AND ( ( ( B_Parvin ) ) ) )
PLC_B = ( ( Gbg_i AND ( ( ( PLC_B ) ) ) ) AND NOT ( PKA AND ( ( ( NOT Gaq ) ) ) ) ) OR ( Gaq )
Alix = ( ESCRT_III ) OR ( ESCRT_I )
Gas = ( Gbg_s AND ( ( ( Gas ) ) AND ( ( NOT RGS ) ) ) ) OR ( alpha_sR AND ( ( ( NOT Gas ) ) AND ( ( NOT Gbg_s ) ) AND ( ( NOT PKA ) ) ) )
Hsc70 = ( GAK ) OR ( Dynamin )
Tab_12 = ( ( Trafs ) AND NOT ( p38 ) )
Mek = ( ( PAK AND ( ( ( Tpl2 ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Mekk1 AND ( ( ( Raf ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Tpl2 ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Mekk2 AND ( ( ( Raf ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Raf AND ( ( ( Tpl2 ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) ) OR ( ( Mekk3 AND ( ( ( Raf ) ) ) ) AND NOT ( PP2A AND ( ( ( Mek ) ) ) ) )
alpha_sR = ( ( alpha_sR ) AND NOT ( B_Arrestin AND ( ( ( Palpha_sR ) ) OR ( ( NOT alpha_sR AND NOT Palpha_sR AND NOT alpha_sL ) ) ) ) ) OR ( ( alpha_sL ) AND NOT ( B_Arrestin AND ( ( ( Palpha_sR ) ) OR ( ( NOT alpha_sR AND NOT Palpha_sR AND NOT alpha_sL ) ) ) ) ) OR ( ( Palpha_sR AND ( ( ( NOT B_Arrestin ) ) ) ) AND NOT ( B_Arrestin AND ( ( ( Palpha_sR ) ) OR ( ( NOT alpha_sR AND NOT Palpha_sR AND NOT alpha_sL ) ) ) ) )
RalGDS = ( ( ( alpha_sR AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( PKC ) ) AND NOT ( Ras AND ( ( ( PIP3_345 ) ) AND ( ( PDK1 ) ) ) ) ) OR ( ( ( alpha_iR AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( PKC ) ) AND NOT ( Ras AND ( ( ( PIP3_345 ) ) AND ( ( PDK1 ) ) ) ) ) OR ( ( ( alpha_qR AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( PKC ) ) AND NOT ( Ras AND ( ( ( PIP3_345 ) ) AND ( ( PDK1 ) ) ) ) ) OR ( ( ( alpha_1213R AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( PKC ) ) AND NOT ( Ras AND ( ( ( PIP3_345 ) ) AND ( ( PDK1 ) ) ) ) )
MLCP = ( ( ( ( ( ( PKA AND ( ( ( RhoK ) ) ) ) AND NOT ( ILK ) ) AND NOT ( PAK ) ) AND NOT ( Raf ) ) AND NOT ( PKC ) ) ) OR NOT ( PAK OR ILK OR PKC OR PKA OR RhoK OR Raf )
ErbB3_Y1270 = ( EGFR_ErbB3 ) OR ( ErbB2_ErbB3 ) OR ( ErbB3_ErbB4 )
EGFR_T654 = ( PKC )
Mekk3 = ( ( Trafs ) AND NOT ( Gab1 ) ) OR ( ( IL1_TNFR ) AND NOT ( Gab1 ) ) OR ( ( Rac ) AND NOT ( Gab1 ) )
Rabenosyn_5 = ( Rab5 AND ( ( ( PIP3_345 ) ) ) )
MLK2 = ( Rac AND ( ( ( SAPK ) ) ) ) OR ( Cdc42 AND ( ( ( SAPK ) ) ) )
Mekk1 = ( Rho AND ( ( ( Grb2 ) ) OR ( ( Shc ) ) ) ) OR ( NIK AND ( ( ( Shc ) ) OR ( ( Grb2 ) ) ) ) OR ( Grb2 AND ( ( ( Shc ) ) ) ) OR ( Ras ) OR ( Trafs ) OR ( Rac ) OR ( GCK ) OR ( Cdc42 )
RIN = ( Ras )
EGFR_Y920 = ( Src AND ( ( ( EGFR_EGFR_EGF_PM ) ) ) )
ErbB3_Y1241 = ( EGFR_ErbB3 ) OR ( ErbB2_ErbB3 ) OR ( ErbB3_ErbB4 )
ARNO = ( PIP2_45 )
Myosin = ( ( ILK AND ( ( ( NOT MLCP ) ) OR ( ( NOT Myosin ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( PAK AND ( ( ( NOT Myosin ) ) OR ( ( NOT MLCP ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( MLCK AND ( ( ( CaM ) ) AND ( ( NOT MLCP ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( RhoK AND ( ( ( NOT MLCP ) ) OR ( ( NOT Myosin ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( CaM AND ( ( ( MLCK ) ) AND ( ( NOT Myosin ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) ) OR ( ( Myosin AND ( ( ( NOT MLCP ) ) ) ) AND NOT ( MLCP AND ( ( ( Myosin ) ) ) ) )
ErbB4_Free = ( ( ( ( ( ErbB4_Free ) AND NOT ( ErbB2_ErbB4 ) ) AND NOT ( ErbB4_ErbB4 ) ) AND NOT ( ErbB3_ErbB4 ) ) AND NOT ( EGFR_ErbB4 ) ) OR ( ErbB4_Contr )
ErbB3_Y1035 = ( EGFR_ErbB3 ) OR ( ErbB3_ErbB4 ) OR ( ErbB2_ErbB3 )
EGFR_EGFR_TGFa_CCP = ( Eps15 AND ( ( ( EGFR_EGFR_TGFa_PM ) ) AND ( ( PIP2_45 AND Clathrin ) ) AND ( ( Rab5 ) ) ) ) OR ( Epsin AND ( ( ( PIP2_45 AND Clathrin ) ) AND ( ( EGFR_EGFR_TGFa_PM ) ) AND ( ( Rab5 ) ) ) ) OR ( AP2 AND ( ( ( EGFR_EGFR_TGFa_PM ) ) AND ( ( Rab5 ) ) AND ( ( PIP2_45 AND Clathrin ) ) ) ) OR ( ( EGFR_EGFR_TGFa_CCP ) AND NOT ( EGFR_EGFR_TGFa_CCV ) )
EGFR_EGFR_TGFa_End = ( ( EGFR_EGFR_TGFa_End ) AND NOT ( EGFR_Free ) ) OR ( EGFR_EGFR_TGFa_CCV AND ( ( ( Hsc70 AND PIP3_345 AND EEA1 AND Rab5 ) AND ( ( ( GAK OR Rabaptin_5 ) ) ) ) ) )
B_Parvin = ( ILK )
Endophilin = ( Eps15 ) OR ( Epsin ) OR ( Endophilin AND ( ( ( PIP2_45 ) ) ) ) OR ( CIN85 )
PDE4 = ( B_Arrestin AND ( ( ( NOT Erk ) ) ) ) OR ( PKA AND ( ( ( B_Arrestin ) ) ) )
ASK1 = ( Trx )
Mekk2 = ( ( PI3K AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCV OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( Mekk2 ) ) OR ( ( Src AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCV OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( Mekk2 ) ) OR ( ( PLC_g AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCV OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( Mekk2 ) ) OR ( ( Grb2 AND ( ( ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_TGFa_CCP OR EGFR_EGFR_EGF_CCV OR EGFR_EGFR_EGF_CCP OR EGFR_ErbB4 OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_EGF_End ) ) ) ) AND NOT ( Mekk2 ) )
DAG = ( ( PLC_B AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( DGK AND ( ( ( DAG ) ) ) ) ) OR ( ( PLC_g AND ( ( ( PIP2_45 ) ) ) ) AND NOT ( DGK AND ( ( ( DAG ) ) ) ) ) OR ( DAG AND ( ( ( NOT DGK ) ) ) )
AP2 = ( Hip1R ) OR ( PIP3_345 ) OR ( Eps15 ) OR ( Epsin ) OR ( EGFR_EGFR_EGF_PM ) OR ( PIP2_45 ) OR ( EGFR_EGFR_TGFa_PM ) OR ( CIN85 )
Gbg_1213 = ( Ga_1213 ) OR ( alpha_1213R AND ( ( ( NOT Ga_1213 ) ) AND ( ( NOT Gbg_1213 ) ) ) )
Epsin = ( EGFR_Ub ) OR ( PIP2_45 ) OR ( EGFR_EGFR_EGF_PM ) OR ( EGFR_EGFR_TGFa_PM )
Hakai = ( Src AND ( ( ( NOT Ca ) ) AND ( ( E_cadherin ) ) ) )
Sos = ( Crk AND ( ( ( Grb2 ) ) AND ( ( NOT PIP2_45 AND NOT Nck AND NOT Erk ) ) AND ( ( PIP3_345 ) ) ) ) OR ( ( Grb2 AND ( ( ( PIP3_345 ) ) ) ) AND NOT ( Cbl_RTK ) ) OR ( Nck AND ( ( ( Crk ) ) AND ( ( PIP3_345 ) ) ) )
Src = ( ( PTPa AND ( ( ( NOT Src ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( EGFR_Y992 ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( Fak AND ( ( ( PTP1b ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( EGFR_Y1086 ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( Cas AND ( ( ( PTP1b ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( Gas AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( alpha_sR AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( EGFR_Y1148 ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) ) OR ( ( Gai AND ( ( ( B_Arrestin ) ) ) ) AND NOT ( Csk AND ( ( ( Src ) ) ) ) )
ErbB3_Y1178 = ( EGFR_ErbB3 ) OR ( ErbB3_ErbB4 ) OR ( ErbB2_ErbB3 )
EGFR_Y992 = ( ( EGFR_ErbB3 ) AND NOT ( SHP2 AND ( ( ( EGFR_Y992 ) ) ) ) ) OR ( ( EGFR_Y992 ) AND NOT ( SHP2 AND ( ( ( EGFR_Y992 ) ) ) ) ) OR ( ( EGFR_ErbB2 ) AND NOT ( SHP2 AND ( ( ( EGFR_Y992 ) ) ) ) ) OR ( ( EGFR_ErbB4 ) AND NOT ( SHP2 AND ( ( ( EGFR_Y992 ) ) ) ) ) OR ( ( EGFR_EGFR ) AND NOT ( SHP2 AND ( ( ( EGFR_Y992 ) ) ) ) ) OR ( ( EGFR_EGFR_EGF_PM ) AND NOT ( SHP2 AND ( ( ( EGFR_Y992 ) ) ) ) ) OR ( ( EGFR_EGFR_TGFa_PM ) AND NOT ( SHP2 AND ( ( ( EGFR_Y992 ) ) ) ) )
Gaq = ( alpha_qR AND ( ( ( NOT Gaq ) AND ( ( ( NOT Gbg_q ) ) ) ) ) ) OR ( Gaq AND ( ( ( Gbg_q ) ) AND ( ( NOT RGS AND NOT PLC_B ) ) ) )
B_catenin = ( ( Fer AND ( ( ( NOT EGFR_ErbB2 AND NOT EGFR_EGFR_TGFa_CCV AND NOT EGFR_EGFR_TGFa_PM AND NOT EGFR_EGFR_EGF_CCV AND NOT EGFR_ErbB3 AND NOT EGFR_EGFR_TGFa_End AND NOT EGFR_EGFR_EGF_PM AND NOT EGFR_EGFR_EGF_MVB AND NOT EGFR_EGFR_TGFa_CCP AND NOT EGFR_EGFR_EGF_CCP AND NOT Src AND NOT EGFR_ErbB4 AND NOT EGFR_EGFR AND NOT EGFR_EGFR_EGF_End ) ) OR ( ( PTP1b ) ) ) ) OR ( PTP1b AND ( ( ( NOT EGFR_ErbB2 AND NOT EGFR_EGFR_TGFa_CCV AND NOT EGFR_EGFR_TGFa_PM AND NOT EGFR_EGFR_EGF_CCV AND NOT EGFR_ErbB3 AND NOT EGFR_EGFR_TGFa_End AND NOT EGFR_EGFR_EGF_PM AND NOT EGFR_EGFR_EGF_MVB AND NOT EGFR_EGFR_TGFa_CCP AND NOT EGFR_EGFR_EGF_CCP AND NOT Src AND NOT EGFR_ErbB4 AND NOT EGFR_EGFR AND NOT EGFR_EGFR_EGF_End ) ) ) ) ) OR NOT ( EGFR_ErbB2 OR EGFR_EGFR_TGFa_CCV OR PTP1b OR EGFR_EGFR_TGFa_PM OR EGFR_EGFR_EGF_CCV OR EGFR_ErbB3 OR EGFR_EGFR_TGFa_End OR EGFR_EGFR_EGF_PM OR EGFR_EGFR_EGF_MVB OR EGFR_EGFR_TGFa_CCP OR Fer OR EGFR_EGFR_EGF_CCP OR Src OR EGFR_ErbB4 OR EGFR_EGFR OR EGFR_EGFR_EGF_End )
EGFR_EGFR_EGF_PM = ( ( EGFR_EGFR_EGF_PM ) AND NOT ( EGFR_EGFR_EGF_CCP ) ) OR ( ( ( EGFR_Free AND ( ( ( EGF ) ) AND ( ( NOT EGFR_T654 ) ) ) ) AND NOT ( EGFR_EGFR_EGF_CCP ) ) AND NOT ( ErbB2_Free ) )
EGFR_EGFR_EGF_End = ( ( EGFR_EGFR_EGF_End ) AND NOT ( EGFR_EGFR_EGF_MVB ) ) OR ( ( EGFR_EGFR_EGF_CCV AND ( ( ( Hsc70 AND GAK AND PIP3_345 AND Rabaptin_5 AND EEA1 AND Rab5 ) ) ) ) AND NOT ( EGFR_EGFR_EGF_MVB ) )
ErbB3_Y1180 = ( EGFR_ErbB3 ) OR ( ErbB2_ErbB3 ) OR ( ErbB3_ErbB4 )
PI3K = ( ( Gbg_i ) AND NOT ( PI3K_I ) ) OR ( ( Fak ) AND NOT ( PI3K_I ) ) OR ( ( E_cadherin ) AND NOT ( PI3K_I ) ) OR ( ( ErbB3_Y1257 ) AND NOT ( PI3K_I ) ) OR ( ( Ras ) AND NOT ( PI3K_I ) ) OR ( ( Src AND ( ( ( Cbl_RTK ) ) ) ) AND NOT ( PI3K_I ) ) OR ( ( EGFR_Y920 ) AND NOT ( PI3K_I ) ) OR ( ( ErbB4_Y1056 ) AND NOT ( PI3K_I ) ) OR ( ( ErbB3_Y1203_05 ) AND NOT ( PI3K_I ) ) OR ( ( Gab1 ) AND NOT ( PI3K_I ) ) OR ( ( ErbB3_Y1270 ) AND NOT ( PI3K_I ) ) OR ( ( EGFR_Y845 ) AND NOT ( PI3K_I ) ) OR ( ( Crk ) AND NOT ( PI3K_I ) ) OR ( ( ErbB3_Y1178 ) AND NOT ( PI3K_I ) ) OR ( ( ErbB3_Y1035 ) AND NOT ( PI3K_I ) ) OR ( ( ErbB3_Y1241 ) AND NOT ( PI3K_I ) )
|
Integrative Modelling of the Influence of MAPK Network
on Cancer Cell Fate Decision
Luca Grieco1,2,3,4,5,6*, Laurence Calzone6,7,8, Isabelle Bernard-Pierrot6,9, Franc¸ois Radvanyi6,9,
Brigitte Kahn-Perle`s2, Denis Thieffry2,3,4,5,10*
1 Aix-Marseille Universite´, Marseille, France, 2 TAGC – Inserm U1090, Marseille, France, 3 Institut de Biologie de l’Ecole Normale Supe´rieure (IBENS), Paris, France, 4 UMR
8197 Centre National de la Recherche Scientifique (CNRS), Paris, France, 5 Inserm 1024, Paris, France, 6 Institut Curie, Paris, France, 7 Inserm U900, Paris, France, 8 Ecole des
Mines ParisTech, Paris, France, 9 UMR 144 Centre National de la Recherche Scientifique (CNRS), Paris, France, 10 INRIA Paris-Rocquencourt, Rocquencourt, France
Abstract
The Mitogen-Activated Protein Kinase (MAPK) network consists of tightly interconnected signalling pathways involved in
diverse cellular processes, such as cell cycle, survival, apoptosis and differentiation. Although several studies reported the
involvement of these signalling cascades in cancer deregulations, the precise mechanisms underlying their influence on the
balance between cell proliferation and cell death (cell fate decision) in pathological circumstances remain elusive. Based on
an extensive analysis of published data, we have built a comprehensive and generic reaction map for the MAPK signalling
network, using CellDesigner software. In order to explore the MAPK responses to different stimuli and better understand
their contributions to cell fate decision, we have considered the most crucial components and interactions and encoded
them into a logical model, using the software GINsim. Our logical model analysis particularly focuses on urinary bladder
cancer, where MAPK network deregulations have often been associated with specific phenotypes. To cope with the
combinatorial explosion of the number of states, we have applied novel algorithms for model reduction and for the
compression of state transition graphs, both implemented into the software GINsim. The results of systematic simulations
for different signal combinations and network perturbations were found globally coherent with published data. In silico
experiments further enabled us to delineate the roles of specific components, cross-talks and regulatory feedbacks in cell
fate decision. Finally, tentative proliferative or anti-proliferative mechanisms can be connected with established bladder
cancer deregulations, namely Epidermal Growth Factor Receptor (EGFR) over-expression and Fibroblast Growth Factor
Receptor 3 (FGFR3) activating mutations.
Citation: Grieco L, Calzone L, Bernard-Pierrot I, Radvanyi F, Kahn-Perle`s B, et al. (2013) Integrative Modelling of the Influence of MAPK Network on Cancer Cell
Fate Decision. PLoS Comput Biol 9(10): e1003286. doi:10.1371/journal.pcbi.1003286
Editor: Satoru Miyano, University of Tokyo, Japan
Received January 16, 2013; Accepted September 2, 2013; Published October 24, 2013
Copyright: 2013 Grieco et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: The research leading to these results has received funding from the European Community’s Seventh Framework Programme (FP7/2007-2013) under
grant agreement nu HEALTH-F4-2007-200767 for APO-SYS(http://cordis.europa.eu/fp7/). It has been further supported by the Agence Nationale de la Recherche,
France (project MI2 iSA). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: grieco@tagc.univ-mrs.fr (LG); thieffry@ens.fr (DT)
Introduction
Mitogen-activated protein kinase (MAPK) cascades can be
activated by a wide variety of stimuli, such as growth factors and
environmental stresses. They affect diverse cellular activities,
including gene expression, cell cycle machinery, apoptosis and
differentiation.
A recurrent feature of a MAPK cascade is a central three-tiered
core signalling module, consisting of a set of sequentially acting
kinases. MAPK kinase kinases (MAPKKKs) are activated follow-
ing upstream signals. For instance, they can be phosphorylated by
small G-proteins belonging to the Ras/Rho family in response to
extracellular stimuli. Their activation leads to double phosphor-
ylation and activation of downstream MAPK kinases (MAPKKs),
which in turn double phosphorylate MAPKs. Once activated,
MAPKs act on their target substrates, which include other kinases
and transcription factors [1]. To date, three main cascades have
been extensively studied, named after their specific MAPK
components: Extracellular Regulated Kinases (ERK), Jun NH2
Terminal Kinases (JNK), and p38 Kinases (p38). These cascades
are strongly interconnected, forming a complex molecular network
[1–4].
MAPK phosphorylation level is regulated by the opposing
actions of phosphatases. As the effects of MAPK signalling have
been shown to depend on the magnitude and duration of kinase
activation, phosphatase action might play an important functional
role [5]. Moreover, scaffold proteins bring together the compo-
nents of a MAPK cascade and protect them from activation by
irrelevant stimuli, as well as from negative regulators (such as
phosphatases) [6].
The involvement of MAPK cascades in major cellular processes
has been widely documented [1,7,8]. However, the wide range of
stimuli and the large number of processes regulated, coupled with
the complexity of the network, raises the fundamental and debated
question of how MAPK signalling specificity is achieved [9].
Several interrelated mechanisms have been proposed: opposing
action of phosphatases; presence of multiple components with
different roles at each level of the cascade (e.g. different isoforms of
a protein); interaction with scaffold proteins; distinct sub-cellular
localisations of cascade components and/or targets; feedback
PLOS Computational Biology | www.ploscompbiol.org
1
October 2013 | Volume 9 | Issue 10 | e1003286
mechanisms; great variety of molecular signals, as well as distinct
durations and strengths; cross-talks among signalling cascades that
are activated simultaneously [4,10]. All these factors contribute to
the complexity of the MAPK network and presumably act
together to determine signalling specificity.
Deregulations of the MAPK cascades are often observed in
cancer [11,12]. Several components of the network have already
been proposed as targets in cancer therapy, such as p38, JNK,
ERK, MEK, RAF, RAS, and DUSP1 [12–23], but the intricacy of
the underlying mechanisms still hinders the conception of effective
drugs [24]. A deeper understanding of the regulatory mechanisms
involved is crucial to clarify the roles of MAPKs in cancer onset
and development, as well as to delineate therapeutic strategies.
During the last decades, mathematical modelling has been
widely used to study different aspects of the MAPK cascades [25]
(Table S1). Quantitative models (based on Ordinary Differential
Equations – ODE) have been proposed to explain the main
dynamical properties of the MAPK cascades in relation with their
particular structural features (double phosphorylation events,
phosphatase effects, feedback loops, role of scaffold proteins,
etc.) [26–32]. Other modelling studies investigated the behaviour
of specific cascades (mainly ERK) leading to MAPK activation in
response to external stimuli [33–42]. More recently, comprehen-
sive qualitative dynamical models have been developed. Samaga et
al. [43] built a large logical model of the signalling network
(including MAPKs) responding to Epidermal Growth Factor
Receptor (EGFR) stimuli, which was largely derived from the
reaction map published by Oda et al. [44]. This model accurately
covers the early response of the MAPK cascades to signalling
stimuli, with a particular reference to primary and transformed
hepatocytes. Also focusing on cancer (in particular, epithelial
tumours), the logical model proposed by Poltz and Naumann
recapitulates the response of a cell to DNA damaging agents (DNA
repair versus apoptotic cell death), and was used to identify
candidate target molecules to design novel therapies [45].
In this study, we aimed at better understanding how MAPK
signalling deregulations can interfere with tissue homeostasis,
leading to imbalance between cell proliferation, on the one hand,
and cell growth arrest, possibly followed by apoptotic cell death,
on the other hand. The choice between these phenotypes (cell fate
decision) is of vital importance in cancer progression: transformed
cells receive external and/or autocrine growth stimuli pushing
towards cell proliferation (i.e. tumour growth); but they also
receive external and/or internal anti-proliferative signals, which
coupled with apoptotic stimuli trigger transformed cell clearance
from the organism [46]. Our goal was to build a predictive
dynamical model able (i) to recapitulate the response of the entire
MAPK network to selected stimuli, along with its specific
contribution to cell fate decision, and (ii) to assess novel hypotheses
about poorly documented mechanisms involved in specific cancer
cell types. We focused on urinary bladder cancer, where MAPK
network deregulations have been associated with specific pheno-
types.
Bladder cancer is the fourth most common cancer among men
in Europe and America. Two main types of early stage bladder
carcinoma have been delineated so far: (i) non-invasive papillary
carcinomas (Ta) are usually mildly aggressive and rarely progress
towards higher stages, whereas (ii) carcinomas in situ (Cis) often
develop into invasive tumours (T1 to T4 stages) [47]. Activating
mutations of Fibroblast Growth Factor Receptor 3 (FGFR3) have
been found in 70–75% of Ta tumours, but they are absent in Cis
and
less
frequent
(15–20%)
in
invasive
tumours
[47,48].
Oncogenic
FGFR3
gene
fusions
have
also
been
recently
identified in a small percentage of invasive bladder tumours
[49]. In contrast, over-expression of EGFR has been recurrently
associated with higher probability of tumour progression [50].
The mechanisms underlying the cellular response of cancer cells
to these signalling stimuli are still poorly understood. Alterations
of p53 and retinoblastoma (RB) pathways are presumably
involved in tumour progression [51]. These pathways are major
controllers of the cell cycle, and the MAPK network presumably
regulates their activation by responding to growth factor stimuli.
For instance, ERK phosphorylation leads to MYC activation,
which can inhibit cell cycle progression through the p14/p53
pathway [52], or activate Cyclin/CDK complexes leading to RB
phosphorylation and subsequent E2F-dependent gene transcrip-
tion [51].
Both EGFR and FGFR3 pathways can activate the MAPK
cascades. Although the two signalling pathways largely overlap,
the presence of specificity factors might contribute to tune the final
cellular response. To tackle these questions, we first compiled
published data to build a comprehensive generic reaction map
using CellDesigner software [53–55]. This map takes into account
signals propagating from major stimuli, such as growth factors,
cytokines, stress, leading to the activation of MAPKs and their
downstream targets, and consequently to cell fate decision. We
considered three main cell fates: proliferation, apoptosis, growth
arrest.
Next, we used the resulting reaction map to design a
qualitative dynamical model with GINsim software [56,57],
which relies on a logical formalism [58–60]. To cope with the
relatively high number of components, we applied a semi-
automatic model reduction procedure to lower the computa-
tional cost of dynamical analyses, without losing the main
dynamical properties of the system. We then performed logical
simulations to check the behaviour of the model in specific
documented situations, as well as to predict the behaviour in
novel situations. We further investigated the role of positive and
negative regulatory circuits in cell fate decision. Altogether, these
analyses provided novel insights into the mechanisms differen-
tiating the response of urinary bladder cancer cells to EGFR
versus FGFR3 stimuli.
Author Summary
Depending on environmental conditions, strongly inter-
twined cellular signalling pathways are activated, involving
activation/inactivation of proteins and genes in response
to external and/or internal stimuli. Alterations of some
components of these pathways can lead to wrong cell
behaviours. For instance, cancer-related deregulations lead
to high proliferation of malignant cells enabling sustained
tumour growth. Understanding the precise mechanisms
underlying these pathways is necessary to delineate
efficient therapeutical approaches for each specific tumour
type. We particularly focused on the Mitogen-Activated
Protein Kinase (MAPK) signalling network, whose involve-
ment in cancer is well established, although the precise
conditions leading to its positive or negative influence on
cell proliferation are still poorly understood. We tackled
this problem by first collecting sparse published biological
information into a comprehensive map describing the
MAPK network in terms of stylised chemical reactions. This
information source was then used to build a dynamical
Boolean
model
recapitulating
network
responses
to
characteristic stimuli observed in selected bladder cancers.
Systematic model simulations further allowed us to link
specific network components and interactions with prolif-
erative/anti-proliferative cell responses.
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
2
October 2013 | Volume 9 | Issue 10 | e1003286
Methods
Logical modelling
We built our dynamical model using the logical formalism
originally proposed by Thomas [58,59]. Implemented in GINsim,
our logical modelling approach initially requires the delineation of
a regulatory graph, where each vertex (node) represents a model
component and each arc (signed, directed edge) represents an
interaction (activation or inhibition) between two components.
Here, all components are associated with Boolean variables,
meaning that they can take two possible levels, 0 or 1, denoting the
absence/inactivation or the presence/activition of the modelled
entities (protein activation level, gene expression level, activation of
a cellular process, etc.). The model definition is completed by
assigning a logical rule to each component. This logical rule
specifies the target value of the component depending on the
presence/absence of its regulators, using the classical Boolean
operators AND, OR and NOT.
Logical simulations
The dynamical behaviour of the model can be computed
starting from any initial state, step by step, updating the current
state according to the logical formulae (logical simulations) [60].
Updating policy.
Two updating policies are mainly used.
According to the synchronous policy, all components are updated
simultaneously at each step; consequently, each state has at most
one successor. In contrast, according to the asynchronous policy,
only one variable can be updated at each step and all the possible
successors of a state are computed. Mixed policies based on the
notion of priority classes can also be defined using GINsim [61],
where subsets of components are ranked. At each step, highest
rank variables are then updated in a synchronous or asynchronous
way.
In this work, we have used the fully asynchronous updating
policy, which usually generates more realistic behaviours [58].
State transition graphs and attractors.
The dynamics of a
logical model can be represented in terms of a state transition
graph (STG), in which nodes denote different states of the system
(represented by a Boolean vector encompassing the values of all
the components), whereas arcs represent enabled transitions
between pairs of states. Of particular interest are the states
forming attractors, i.e. (groups of) states from which the system
cannot escape, which represent potential asymptotical behaviours.
Attractors can be ranged into two main classes:
–
stable states, corresponding to fixed points (i.e. states without
successors);
–
cyclic attractors, corresponding to terminal cycles or to more
complex terminal strongly connected components, comprising
several intertwined cycles.
Leaning on a representation of the logical rules in terms of
Multi-valued Decision Diagrams (MDD), an algorithm enables the
computation of all the stable states of a logical model (indepen-
dently of the initial conditions) [62]. The efficiency of the
algorithm (which does not require to compute the state transition
graph) makes this tool particularly useful when dealing with large
logical models. However, other means are needed to assess the
reachability of the stable states from specific initial states, or yet to
identify cyclic attractors.
Deeper dynamical analyses imply the computation of the state
transition graph. GINsim user can define a set of initial states and
an updating strategy; the software then computes the state
transition graph, highlighting stable states and cyclic attractors.
GINsim further eases the definition of perturbations, which are
simulated by forcing the level of a subset of components to fixed
values (or value intervals). For instance, in the Boolean case, we
can reproduce a loss-of-function by setting a component to 0,
whatever the levels of its regulators, whereas a gain-of-function can
be simulated by forcing the corresponding component to 1. More
subtle perturbations can be simulated by rewriting relevant logical
rules.
As the size of the model considered increases, we are facing the
well known problem of the exponential growth of the state
transition graph. To cope with this problem, we used two methods
that amount to compress the model before simulation or to
compress the resulting state transition graph on the fly. These two
methods are briefly described hereafter, along with a complemen-
tary method enabling the identification of regulatory circuits
playing crucial dynamical roles.
Model reduction
To deal with large models, GINsim enables their reduction by
‘‘hiding’’ selected components [63]. In practice, the user selects the
components to hide, and the software hides each of them
iteratively, while recomputing the logical rules of their targets.
Provided that no functional regulatory circuit is eliminated in the
process, this reduction preserves all attractors. For example, the
stable states are all conserved, in the sense that each stable state of
the reduced model is the projection (on the reduced state space) of
a stable state of the original model [63]. This tool is particularly
useful when the high dimensionality impedes the computation of
the full STG.
Hierarchical transition graph representation
The analysis of the paths from initial states to attractors can be
done directly on the STG when it is small (tens of states), but
becomes quickly intractable as the size of STG increases. To cope
with this difficulty, we use a novel feature of GINsim, which
iteratively clusters the state transition graph into groups of states
(components or hyper-nodes) sharing the same set of successors
[64]. The resulting hierarchical state transition graph (HTG)
displays all the reachable attractors (components at the bottom of
the graph), while the other clusters of states represent their basins
of attractions (including strict basins with a single outgoing arc
targeting a specific attractor, or non-strict basins grouping states
that can reach a specific set of HTG components). HTG
computation is done on the fly, i.e. without having to store the
whole STG, which often leads to strong memory and CPU usage
shrinking. Furthermore, this functionality eases the identification
of the key commutations (changes of component levels) underlying
irreversible choices between the different reachable attractors.
Altogether, the HTG representation is very compact (often much
more compact than the more classical graph of strongly connected
components, as HTG further compacts linear/non circular
pathways) and very informative regarding the organisation of the
original STG.
Regulatory circuit analysis
Rene´ Thomas proposed generic rules linking the presence of
regulatory circuits (simple oriented regulatory cycles) in biological
networks with dynamical properties. The first rule states that the
existence of a positive circuit (involving an even number of
negative regulatory interactions) is a necessary condition for multi-
stationarity. The second rule states that the existence of a negative
regulatory circuit (involving an odd number of negative regulatory
interactions) is a necessary condition for the generation of
sustained oscillations [65]. More recently, Remy et al. [66]
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
3
October 2013 | Volume 9 | Issue 10 | e1003286
translated these rules into theorems in the case of asynchronous
Boolean dynamical systems (which is the case of our MAPK
model).
However, when embedded in a more complex network, specific
constraints on the values of the external components acting on
circuit components have to be fulfilled in order to allow a
regulatory circuit to produce the expected behaviour (‘‘circuit
functionality constraints’’) [67]. The concept of circuit function-
ality has been formalised for logical models and implemented into
GINsim [62].
GINsim allows to compute all the functional positive and
negative circuits of a model. For each of them, the software also
provides the corresponding functionality context, defined as a set
of constraints on the values of its external regulators. This tool
enables the identification of the regulatory circuits playing key
dynamical roles within a complex network.
Results
MAPK reaction map
Based on published data, we have built and annotated a
comprehensive reaction map using CellDesigner (supplementary
Dataset S1). This map encompasses 232 species (proteins, genes,
complexes, other molecules) and 167 reactions involved in the
three most extensively documented MAPK cascades (ERK, JNK,
p38). The CellDesigner version of the map (XML format) is
provided as supplementary Dataset S2. The MAPK map has been
further integrated into the Atlas of Cancer Signalling Networks
developed by the group of Emmanuel Barillot at Institut Curie in
Paris (https://acsn.curie.fr), where it can be explored using a web
browser.
Our reaction map takes into account signals propagating from
different major stimuli, such as growth factors, cytokines, stress,
which lead to the activation of MAPKs and their downstream
targets. It covers feedbacks and cross-talks among the MAPK
cascades, as well as the roles of the best documented phosphatases
and scaffold proteins. The main cellular compartments are also
represented (plasma membrane, cytoplasm, nucleus, mitochon-
dria, endosomes, etc.), showing the localisation of reactions within
the cell. When compartmentalisation has not been fully char-
acterised yet, reactions have been provisionally assigned to the
cytoplasm. Proteins are coloured to emphasise relevant families.
Figure 1 shows a map excerpt reporting two different mechanisms
of ERK activation (see map annotations for more details).
We
considered
two
compartments
named
‘‘Genes’’
and
‘‘Phenotypes’’
at
the
bottom
of
the
map,
which
include
representative genes activated by the MAPK cascades, as well as
phenotypes (proliferation, apoptosis, growth arrest) enabled by
selected readouts.
We considered information concerning different human and
mouse cell types, implying that the MAPK map should be
considered as generic. Indeed, at this stage, information is lacking
to build a detailed map based exclusively on data for a specific cell
type. However, we selected biological events explicitly considered
to be cell type independent. When applicable, information
concerning cell types is provided through links to relevant
database entries (mainly PubMed).
Because the precision of the information retrieved from the
literature varies, our map represents different pathways with
different levels of details. For instance, we could find detailed
information about the scaffold proteins intervening in the ERK
cascade and on the sub-cellular localisation of the correspond-
ing protein complexes; in contrast, such information is still
largely lacking for the JNK and p38 cascades. This is why the
map currently reaches its greatest level of detail for the ERK
cascade.
Furthermore, the level of detail represented could also be
dictated by readability considerations. For instance, the RTK
(receptor tyrosine kinase) component in the map represents several
different receptors (e.g. EGFR, FGFR, VEGFR, etc.): all these
receptors share common features that are related to MAPK
activation. However, their mechanisms of action may differ in
some subtle ways, which are not fundamental for our purpose
here. The detailed representation of all these pathways would have
made the map very difficult to read, and we thus decided to
simplify the graphical representation, while keeping track of
relevant variations in the annotations of the corresponding species
or reactions.
The resulting CellDesigner map constitutes a comprehensive
and integrated source of information concerning the roles of the
MAPK network in cell fate decision, taking into account specificity
factors. This map can be directly used by biologists and modellers
to get information about the reported phenomena. It can also be
used for visualisation of high-throughput data (e.g. by automat-
ically colouring components based on expression levels) derived
from different cell conditions, for example in order to identify
differentially expressed components. This can also give insights
into cell type-dependent mechanisms.
MAPK logical model
Hereafter, we focus on the impact of the MAPK network in
urinary bladder cancer, with particular emphasis on the differen-
tial behaviour between EGFR over-expression and FGFR3
activating mutation.
Scope of the MAPK logical model.
In order to study the
response of the MAPK network to specific stimuli, and its
influence on cell fate decision, we built a dynamical model
covering the mechanisms reported in the MAPK map. We derived
a regulatory graph using the MAPK reaction map as an
information source, as detailed in supplementary Text S1. In
particular, we considered the following subset of stimuli in our
model: EGFR stimulus, FGFR3 stimulus, TGFbR stimulus, and
DNA damage. With reference to the latter stimulus, please notice
that we did not consider explicitly here the DNA repair process
following DNA damage, but we only account for the triggering of
growth arrest and apoptosis following sustained DNA damage
[68]. In our dynamical model, DNA damage thus corresponds to
sustained stress or to the effects of therapies involving DNA-
damaging agents.
The regulatory graph shown in Figure 2 covers the activation of
MAPK targets that influence the choice between proliferation,
growth arrest, and apoptosis. In particular, we consider MYC and
p70 (in the absence of p21) as markers of cell cycle enablement,
p21 as a marker of growth arrest, FOXO3 and p53 as markers of
apoptosis,
whereas
ERK
and/or
BCL2
indicate
apoptosis
disablement. To ease the interpretation of phenotypes, we defined
three output nodes denoting proliferation, growth arrest, and
apoptosis,
respectively.
These
nodes
represent
enablement/
disablement of the corresponding processes, depending on the
MAPK network state, but not necessarily all requirements for this
phenotype. For instance, when proliferation is enabled (by the
interplay of MYC, p70 and p21), we assume that Cyclin/CDK
complexes are activated. In this context, the node ‘‘Proliferation’’
denotes entry into the cell cycle and does not account for
alterations of later stages of the cell cycle. Similar considerations
are applicable for the other phenotypes modelled. This simplifi-
cation is justified by our focus on the specific contributions of the
MAPK network to cell fate decision.
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
4
October 2013 | Volume 9 | Issue 10 | e1003286
Each of the 52 components of the regulatory graph is modelled
by a Boolean variable along with a logical rule (Table S2)
specifying how the component activity depends on its regulators.
Reduced model versions.
To cope with the relatively high
dimension (52 components) of our model, we took advantage of
the model reduction function implemented in GINsim (see
Methods). Indeed, it is difficult to perform simulations with the
original model version, as it entails 252 states. The choice of
components to hide depends on the simulation performed. For
instance, if we plan to simulate a p53 loss-of-function and observe
its effects on p21, we better conserve p53 (otherwise we cannot
define the perturbed version) and p21 (otherwise, we cannot
explicitly observe its value). However, as we wanted to test several
situations, almost half of the MAPK model components were
needed to be observable. Consequently, we designed several
reduced versions of the original MAPK model, each of which
dedicated to a subset of simulations. This strategy enabled us to
drastically reduce the computational cost of our in silico
experiments (the dimension of the reduced models ranged from
16 to 18 components).
Altogether, we defined three different reduced model versions,
whose component lists are reported in supplementary Table S3.
These definitions are enclosed in the comprehensive model file
(supplementary Dataset S4) and enable the generation of reduced
versions according to simulation needs. Figure 3 shows the
regulatory graph corresponding to one reduced version, namely,
‘‘red1’’. Supplementary Dataset S3 lists the simulations performed
for each reduced version. The three model reductions were found
equivalent in terms of attractors and main dynamical properties.
Briefly, reduction ‘‘red1’’ was used to obtain the results discussed
in the sections ‘‘Coherence with well established bladder cancer
deregulations’’ and ‘‘Predictions generated with the MAPK logical
model’’, while reductions ‘‘red2’’ and ‘‘red3’’ were used to obtain
the results discussed in the section ‘‘Coherence with additional
cancer-related facts’’.
Coherence of the logical model behaviour with
published data
The logical rules assigned to model components were inferred
from information about a broad range of experiments and cellular
conditions. To check the coherence of the global behaviour of the
resulting model with current biological knowledge, we systemat-
ically compared its dynamical properties with published data
concerning different tumoural cell types, with particular attention
to bladder cancer. More specifically, we first assessed the
dynamical
behaviour
of the
model under
well established
perturbations observed in the bladder cancer subtypes of interest.
We further checked the coherence of the model behaviour with an
additional list of biological facts, not necessarily involved in
bladder cancer. These analyses were carried out by performing
asynchronous simulations for selected initial conditions (initial
states, input signals, potentially in the presence of perturbations),
and observing the attractors reached by the system.
Figure 1. Molecular map for ERK regulation and sub-cellular localisation. After RAS activation, ERK cascade can be recruited and activated
on plasma membrane with the help of KRS1 scaffold protein (upper part of the figure); activated ERK is then released (in complex with MEK and KSR1)
into the cytoplasm, where it can activate some of its cytoplasmic targets (e.g. PLA2G4A protein). Alternatively, activated receptor complex can
translocate to late endosomes (left part of the figure), where ERK cascade can be triggered with the help of MP1 scaffold protein; in this case,
activated ERK monomers are released into the cytoplasm, from where they can translocate into the nucleus and exert other effects (e.g. induction of
DUSP1 phosphatase). This map is a small fraction of the detailed MAPK network built with the software CellDesigner (www.celldesigner.org) and
provided in png and cell formats (supplementary Datasets S1 and S2).
doi:10.1371/journal.pcbi.1003286.g001
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
5
October 2013 | Volume 9 | Issue 10 | e1003286
In practice, the entire process from reaction map construction
to model simulations is iterative, requiring several rounds of
literature searches and in silico experiments. Whenever the model
disagreed with established facts, we went back to the literature to
seek complementary information and refined our modelling
hypotheses. The reaction map and the logical model where
systematically and consistently completed with relevant informa-
tion during this process.
Coherence
with
well
established
bladder
cancer
deregulations.
As we build our model around the comparison
between EGFR over-expression and FGFR3 activating mutation
in bladder cancer, our first simulations were dedicated to the
assessment of the model behaviour in these circumstances.
Figure 4a reports a simplified view of the model dynamics
following EGFR over-expression, obtained by setting EGFR to 1
throughout the simulation, in the absence of additional stimuli (see
supplementary Dataset S3 for precise simulation settings). In
response to EGFR over-expression, the asynchronous state
transition
graph
encompasses
two
sets of
trajectories:
one
characterised by p53 activation and ERK silencing, leading to
an apoptotic stable state (i.e. Apoptosis = Growth_Arrest = 1,
Proliferation = 0); the other characterised by p53 silencing and
ERK activation, leading to a PI3K/AKT-dependent proliferation
stable state (i.e. Apoptosis = Growth_Arrest = 0, Proliferation = 1)
(cf. simulation provided in the supplementary Text S2).
Alterations in the p53 pathway have been associated with more
aggressive and invasive bladder cancers [50]. The fundamental
role of p53 in the model can be observed by simulating a p53
loss-of-function (second row of Figure 4c). In this case, the
proliferative attractor is kept, while the apoptotic attractor is lost.
In contrast, still in presence of EGFR over-expression, when the
system is also subjected to persistent DNA damage (third row of
Figure 4c), we obtain a single apoptotic attractor. This behaviour
is in agreement with the fact that when damage is moderate (i.e.
absence of DNA damage stimulus), the cell is still able to escape
apoptotic cell death by down-regulating p53 signalling (i.e.
possible switch between the two attractors of Figure 4a), but p53
eventually induces apoptosis in cells subjected to extensive DNA
damage [69].
A similar response is also predicted in the case of TGFBR
stimulus, in line with the typical anti-proliferative role played by
this pathway [70]. In this respect, TGFBR has also been shown to
induce proliferation in tumours, in some circumstances, but the
conditions (especially in relation with MAPK network) under
which this occurs are still poorly understood [70]. Strong
activation of PI3K/AKT pathway has also been associated with
enhanced bladder tumour proliferation [50]. Accordingly, in our
model, PI3K/AKT gain-of-functions counteract p53 pathway
effects (fifth row of Figure 4c), impairing the apoptotic phenotype.
Two other important loss-of-function known to be associated
with poorer prognosis in bladder cancers have been simulated.
Deletions of either PTEN or p14 in our model are associated with
a more aggressive phenotype (sixth and seventh row of Figure 4c).
The former is a tumour suppressor shown to inhibit AKT
Figure 2. Regulatory graph of the MAPK logical model. Each node denotes a model component. Model inputs, phenotypes and MAPK
proteins (ERK, p38, JNK) are denoted in pink, blue and orange, respectively. Green arrows and red T-arrows denote positive and negative regulations,
respectively. A comprehensive documentation is provided in the Table S4, which includes a summary of all modelling assumptions, references
(PubMed links) and the specification of the logical rule associated with each component. The source file is further provided as supplementary Dataset
S4, which can be opened, edited, analysed and simulated with the softare GINsim (www://www.ginsim.org/beta).
doi:10.1371/journal.pcbi.1003286.g002
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
6
October 2013 | Volume 9 | Issue 10 | e1003286
expression [71]. The latter is induced by MYC and is able to
enhance p53 activity by inhibiting MDM2 [51]. According to our
model, p14 loss-of-function abrogates apoptosis, which accounts
for the observed dual role of MYC [72]: on the one hand, MYC
contributes to proliferation (MYC is a read-out for proliferation in
the model); on the other hand, it is involved in p53-dependent
apoptosis.
The behaviour of our model in the case of FGFR3 activating
mutation is depicted in Figure 4b (cf. Text S2 for a complete
simulation). We find again the ‘‘p53 versus ERK’’ pattern
accounting for the fundamental role of p53 in cell fate decision.
Interestingly, the non-apoptotic branch of the asynchronous state
transition graph is now characterised by two different behaviours.
On the one hand, similarly to EGFR over-expression, when PI3K
is active, the system will eventually reach a proliferative attractor.
On the other hand, a p53-independent PI3K/AKT pathway leads
to an attractor characterised by all phenotypes set to 0, that we
interpret as ‘‘no cell fate decision taken’’ at the level of MAPK
network. This is coherent with the contention that FGFR3
mutations, mainly characterising non-invasive bladder carcino-
mas, relatively mildly induce proliferation due to the action of
ERK cascade [47,51,73]. Indeed, the coexistence of these
outcomes (proliferation versus no cell fate decision) tentatively
explain the less aggressive phenotype generally observed in
FGFR3-mutated bladder carcinomas, in comparison with urothe-
lial tumours over-expressing EGFR (associated only with a
proliferative attractor). The underlying mechanisms are further
analysed below. By and large, the simulations of FGFR3 mutation
correctly recapitulate the effects of the major bladder cancer
deregulations listed in Figure 4c (left column), producing results
that are qualitatively coherent with those described for EGFR
over-expression.
Coherence
with
additional
cancer-related
facts.
To
further assess the consistency of the behaviour of our model with
current knowledge, we selected a list of established facts regarding
the effects of perturbations on (human/mouse) cancer cells, not
necessarily bladder-related. Based on this list, we defined a series of
in silico experiments, combining relevant initial states and virtual
perturbations (loss-of-functions and/or gain-of-functions of select-
ed model components, as defined in Methods).
This analysis mainly consisted in cross-checking the attractors
obtained in our simulations with the qualitative information
retrieved from the literature, without focusing on the full dynamics
of the system.
A summary of the results obtained is provided in Table 1 (for
more details, see the supplementary Dataset S3). Additionally, all
the simulations performed can be easily reproduced using the
model files available as supplementary Dataset S4. Briefly, the
involvement of MAPKs in cell fate decision was assessed through
perturbations of relevant components. The model accounts for the
pro-apoptotic role of p38 and JNK, as well as for the promotion of
growth arrest by p38, and for the proliferative role of ERK. The
model also recapitulates the p21-mediated tumour suppressor
function of p53, along with the impairment of this function due to
epigenetic silencing of GADD45. Additionally, we were also able
to reproduce (i) the positive effects of EGFR/FGFR3/RAS/RAF
over-expressions on ERK activation; (ii) the negative effects of
HSP90-inhibitors on cell proliferation; (iii) the role of ERK against
anti-proliferative TGFb signalling; and (iv) the role of JNK against
RAS-induced proliferation. These simulations cover the most
Figure 3. Regulatory graph of a reduced version of the MAPK model. The regulatory graph corresponds to the ‘‘red1’’ reduced model
version (cf. supplementary Table S3, column 1). To obtain this version, the preservation of pink and blue nodes was imposed, along with that of
{EGFR, FGFR3, p53, p14, PI3K, AKT, PTEN, ERK}, in order to investigate the effects of perturbations affecting these components. The remaining nodes
{FRS2, MSK} were maintained by the reduction algorithm because of the occurrence of auto-regulatory loops during the reduction process. Green
arrows and red T-arrows denote positive and negative regulations, respectively, whereas blue arrows denote dual interactions.
doi:10.1371/journal.pcbi.1003286.g003
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
7
October 2013 | Volume 9 | Issue 10 | e1003286
Figure 4. Coherence of the logical model with well established bladder cancer deregulations. a) Simplified representation of the model
dynamics following EGFR over-expression (EGFR = 1 throughout the simulation and all inputs set to 0 throughout the simulation). If p53 is activated
first (right branch), an apoptotic attractor is reached, characterised by inactivation of ERK and AKT. If ERK and PI3K are activated first (left branch), then
p53 is inactivated and AKT is activated, leading to a proliferative attractor. b) Simplified representation of the model dynamics following FGFR3
activating mutation (FGFR3 = 1 and all inputs set to 0 throughout the simulation). When p53 is activated first (right branch), an apoptotic attractor is
reached, characterised by inactivation of ERK and AKT. If ERK is activated first (left and central branches), then p53 is inactivated. When PI3K is also
activated (central), a proliferative attractor is reached, characterised by activated AKT. In contrast, when PI3K is not activated (left), the cell fails to
make a clear decision at the level of the MAPK network. c) Attractors reached by the model in presence of receptor alterations, coupled with
additional common deregulations observed in bladder cancer. Coloured circles denote the phenotypes characterising the attractors reached in each
situation (we used the same colour code as in panels a and b, while empty spaces denote the loss of the corresponding branch in the state transition
graph). Identifiers in rectangles (e.g.. r3, r4, etc.) point to simulation results reported in more details in Dataset S3 and Text S2.
doi:10.1371/journal.pcbi.1003286.g004
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
8
October 2013 | Volume 9 | Issue 10 | e1003286
salient behaviours of the model, showing a remarkable coherence
with published data.
Predictions generated with the MAPK logical model
Having shown that our MAPK model is consistent with
published data, we designed additional simulations to explore
novel mechanistic hypotheses.
ERK-related
feedback
mechanisms.
So far, we have
described the behaviour of our model in presence of tumoural
deregulations of growth factor receptors. Let us consider now what
happens when the expression of such receptors is not altered (i.e.
unperturbed logical rules for either EGFR or FGFR3 variables), in
presence of sustained growth factor stimulation (i.e. either
EGFR_stimulus or FGFR3_stimulus set to 1 throughout the
simulations). The attractor reached from normal EGFR stimulation
is characterised by oscillations (between 0 and 1) of the values of
EGFR, ERK and p53, thus leading to oscillations of phenotype
variables, in particular for ‘‘Proliferation’’ (Dataset S3 – r1). This
behaviour contrasts with the well defined phenotypes obtained
following EGFR over-expression (Figure 4a). It can be interpreted
as the impossibility to obtain sustained activation (or inactivation) of
the considered actors in presence of sustained growth factor stimuli.
In other words, ERK oscillations in our state transition graph
correspond to the transient ERK activation in the ODE-based
model from Orton et al. [38]. Similar results are obtained for
FGFR3 stimulation (Figure 4b and Dataset S3 – r2), in agreement
with literature [74].
The main negative feedbacks underlying such responses are
acting directly on the receptors (Figure 2): one accounting for
GRB2-dependent ubiquitination and degradation of the receptors
Table 1. Coherence of model simulations with published experimental evidence.
Reduction
Simulation
Biological data
Model behaviour
red2
r17, r18
* RAF or RAS over-expressions can lead to constitutive
activation of ERK. [11]
In absence of inputs, constitutive activity of any one among
RAF or RAS can lead to permanent ERK activation,
associated with proliferation.
red2
r19
* HSP90-inhibitor disrupts RAF, AKT and EGFR, leading to
successful cancer treatment [86].
Concomitant RAF, AKT, EGFR deletions abrogate the
proliferative stable states observed in the unperturbed
model, both in the case of EGFR over-expression (obvious –
simulation not performed) and in the case of FGFR3
activating mutation.
red2
r20, r21, r22, r23
* Patients with p53-altered/p21-negative tumors
demonstrated a higher rate of recurrence and worse
survival compared with those with p53-altered/
p21-positive tumors [87].
Following either EGFR over-expression or FGFR3 activating
mutation, concomitant p21 and p53 loss-of-functions
correspond to a phenotype characterised by apoptosis
escape (Apoptosis = Growth_Arrest = 0), with the possibility
to attain proliferation. Association of p53 loss-of-function
and p21 gain-of-function leads to growth arrest attractors,
all characterised by no proliferation.
red3
r25
p38 and JNK play important roles in stress responses,
such as cell cycle arrest and apoptosis [7,69].
In presence of either DNA_damage or TGFBR_stimulus,
growth arrest/apoptosis stable states are all lost in the p38/
JNK-deleted model.
red3
r26
p38 and JNK, especially in the absence of mitogenic
stimuli, have been shown to induce apoptotic cell
death [7,69].
When p38/JNK are constitutively active, apoptotic attractors
(Growth_Arrest = Apoptosis = 1, Proliferation = 0) are
obtained in the absence of other stimuli.
red3
r27
p38 plays its tumour suppressive role by promoting
apoptosis and inhibiting cell cycle progression [11].
Under JNK constitutive activation, p38 loss-of-function
determines loss of apoptotic attractors obtained in r26.
red3
r28
JNK may contribute to the apoptotic elimination of
transformed cells by promoting apoptosis [11,88].
Under p38 constitutive activation and JNK loss-of-function,
all apoptotic attractors obtained in r26 become growth
arrest attractors (Growth_Arrest = 1, Apoptosis = 0,
Proliferation = 0), thus determining loss of apoptotic
attractors obtained in r26.
red3
r29
Epigenetic gene silencing of GADD45 family members
has been frequently observed in several types of human
cancers [69].
In presence of DNA_damage (main GADD45 activator),
Growth_Arrest and Apoptosis components permanently
oscillate when GADD45 is silenced, suggesting less
propensity to cell death. Apoptotic stable states are still
reached in presence of TGFBR_stimulus
red3
r30
ERK increases transcription of the cyclin genes and
facilitates the formation of active Cyc/CDK complexes,
leading to cell proliferation [89].
ERK gain-of-function always leads to proliferative attractors
(Proliferation = 1, Growth_Arrest = Apoptosis = 0), in the
absence of other stimuli.
red3
r31
ERK disrupts the anti-proliferative effects of TGFb [11].
Whereas TGFBR_stimulus leads to an apoptotic stable state
(r24), coupling of TGFBR_stimulus with ERK gain-of-function
only leads to permanent growth arrest (Growth_Arrest = 1,
Apoptosis = 0).
red3
r32
JNK might reduce RAS-dependent tumour formation by
inhibiting proliferation and promoting apoptosis [88].
In absence of other stimuli, JNK constitutive activation
completely abrogates RAS-dependent proliferation
following RAS over-expression (r18). Instead, apoptotic
attractors are always reached.
Asterisks denote facts explicitly related to bladder cancer, whereas unmarked entries correspond to generic or loosely specified mechanisms. Full simulation results can
be found in Dataset S3, with the help of the identifiers provided in the first two columns.
doi:10.1371/journal.pcbi.1003286.t001
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
9
October 2013 | Volume 9 | Issue 10 | e1003286
[75]; the other accounting for PKC-mediated negative effects
[74,76]. Concomitant disruptions of these feedbacks in our model
lead to simulation results equivalent to those obtained with
receptor gain-of-function (cf. Figure 2 and Table S2), whereas
disruption of any other downstream negative feedback does not
qualitatively influence the outcome (data not shown). This is also
in agreement with the results obtained by Orton et al., who
proposed that the degradation of receptors (e.g. rather than SOS
inhibition by RSK) could be the main mechanism determining a
transient activation of ERK pathway in response to growth factors.
Role of Sprouty-mediated feedbacks.
According to our
model simulations (Figure 4a), when EGFR is over-expressed (e.g.
in the presence of an autocrine signal), in the absence of p53
activation, the outcome is proliferation. In contrast, when FGFR3
stimulus is present, two possible outcomes are observed in the
absence of p53: a proliferative stable state and a non-proliferative
stable state, the later with all phenotype variables set to 0.
General hypotheses involving the interplay between the p53,
RB and ERK pathways have been proposed to explain the
different phenotypes experimentally observed in bladder carcino-
mas [50,73], but the precise mechanisms have not been elucidated
yet. A closer analysis of the regulatory graph shown in Figure 2
reveals several feedbacks. Interestingly, ERK exerts a positive
feedback on EGFR but a negative feedback on FGFR3-activated
FRS2, through Sprouty (SPRY in the model). Intuitively, this
suggests that ERK strengthens EGFR stimulus but weakens
FGFR3 stimulus, potentially explaining the different phenotypes
observed. Additionally, GRB2 exerts a negative feedback on
FRS2, which is in turn specifically activated by FGFR3.
Disruption of EGFR activation by SPRY does not play an
important role in the case of EGFR over-expression (which indeed
corresponds to setting EGFR to 1 independently of its regulators).
FRS2 inhibition by SPRY, but not by GRB2, tentatively plays an
important role in the response of the MAPK network to FGFR3
activating mutation. Indeed, disrupting the latter inhibition
(Figure 5) does not affect significantly the model behaviour. On
the contrary, comparison of Figures 4a–b and Figure 5 indicates
that SPRY-dependent inhibition of FRS2 might be the key to
explain the difference between the responses to EGFR and
FGFR3 stimulations (i.e. in the absence of this inhibition, the
model behaves equivalently in the two cases).
To further clarify these mechanisms, we considered the role of
functional positive circuits, which are known to promote multi-
stable behaviours (cf. Methods). According to our model analysis,
the GAB1-PI3K-GAB1 circuit underlies the coexistence of the two
stable states found in the presence of FGFR3 activating mutation,
but in the absence of p53 activation. We thus propose that this
circuit plays a fundamental role in FGFR3 signalling, constituting
a switch between proliferative and non-proliferative phenotypes.
The underlying mechanisms can be further clarified by a careful
analysis of Figure 2. On the one hand, following FGFR3 stimulus,
when PI3K activation is faster/stronger than ERK activation, cell
proliferation is enabled, because PI3K is definitively activated (due
to the action of GAB1-PI3K-GAB1 positive circuit) and can then
inhibit p21 through the PDK1/AKT pathway. ERK is then also
activated, enhancing proliferation together with PI3K. On the
other hand, upon ERK activation (coming from a rapid GRB2
and/or PKC mediated signalling), if the inhibition of GRB2
through the SPRY/FRS2 feedback is faster/stronger than PI3K
activation, then PI3K cannot be activated anymore. In this
scenario, ERK would rather contribute to disable cell prolifera-
tion.
Our model thus predicts that the strength/rapidity of PI3K
activation versus SPRY-mediated ERK negative feedback could
underly the less aggressive phenotypes observed in FGFR3-
mutated bladder carcinomas.
Figure 5. FGFR3 activating mutation and role of SPRY. Simulations were performed under FGFR3 gain-of-function (FGFR3 = 1 and all inputs set
to 0, throughout the simulations). Simplified model dynamics are shown as in Figure 4a–b. Results are shown for the wild type model (red1 model
reduction), as well as for perturbed model versions obtained by disrupting the inhibition of interest.
doi:10.1371/journal.pcbi.1003286.g005
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
10
October 2013 | Volume 9 | Issue 10 | e1003286
MAPK cross-talk mechanisms.
Using our logical model,
we further addressed the roles of cross-talks between the different
MAPK cascades, in particular those involving phosphatases.
We first analysed the negative cross-talks from p38/JNK
cascades towards ERK cascade, which involve MEK inhibition
by AP1 and the phosphatase PPP2CA [2]. In the context of either
EGFR over-expression or FGFR3 activating mutation (Figure 6a),
the disruption of these inhibitions mainly lead to the loss of
apoptotic attractors (compare Figure 6a with Figure 4a–b). The
lost attractors are ‘‘replaced’’ by new attractors characterised by
growth arrest alone. These two cross-talks are thus presumably
important for the triggering of apoptotic responses in the presence
of growth factor receptor alterations. Indeed, in the absence of
such cross-talks, p53 pathway is only able to induce growth arrest,
but
not
apoptosis,
precluding
a
complete
anti-proliferative
response. This is also true in the concomitant presence of DNA
damage stimulus (data not shown).
Finally, we examined the roles of p38 and JNK inhibitions by
DUSP1 [77]. Following receptor (either EGFR or FGFR3)
alteration, disruption of any of these two inhibitions results in a
persistent silencing of ERK and a persistent activation of p53, as
well as an activation of PI3K and an inactivation of AKT, which
ultimately lead the system towards an apoptotic attractor (compare
Figure 6b with Figure 4a–b). DUSP1-mediated cross-talks between
the
MAPK
cascades
thus
tentatively
underly
proliferative
responses in presence of growth factor receptor alterations,
presumably via the inhibition of p53 pathway.
Discussion
We have presented a bottom-up modelling approach to gain
insights into the influence of the complex MAPK signalling
network on cancer cell fate decision. We started by collecting
pieces of information from the literature and assembling them into
a detailed reaction map, which served as source of information for
further dynamical modelling. The resulting map is generic,
meaning that it was drawn by using information coming from
different experimental models.
Based on specific biological questions, our dynamical logical
modelling involved the abstraction of relevant information from
the map and the drawing of a qualitative influence network
(regulatory graph). Next, we assigned consistent logical rules to
each component to enable logical simulations. In order to perform
detailed analyses at reasonable computational costs, we derived
reduced model versions preserving the main dynamical properties
of the original model. The reduced versions can be considered as
further
abstractions
of the MAPK network, explaining its
qualitative behaviour in terms of selected molecular actors.
Despite the fact that we made no use of quantitative data, and
that we finally represented an extremely complex signalling
network through a limited number of Boolean components, we
were able to recapitulate its behaviour for diverse documented
biological conditions. These results
set the background
to
investigate the roles of poorly documented regulatory mechanisms.
In this modelling study, we particularly focused on bladder
cancer. Importantly, our simulations qualitatively recapitulated
salient phenotypic differences observed in invasive versus non-
invasive carcinomas, and allowed us to formulate reasonable
hypotheses concerning the mechanisms determining such differ-
ences. These hypotheses are readily amenable to experimental
validation.
Our MAPK network model can be considered as a module for
the assembly of more comprehensive cancer-related network.
From this point of view, it will be interesting to merge our model
with other logical models implementing other aspects of cell fate
decision, in particular the model proposed by Calzone et al. [78],
Figure 6. Analysis of MAPK cross-talks by disruptions of specific interactions. a) Effects of the disruptions of the inhibitions of MEK by AP1
and the phosphatase PPP2CA. b) Effects of the disruption of the inhibition of p38 or of JNK by the phosphatase DUSP1. These simulations were
performed after removing the corresponding interactions and blocking the level of the perturbed receptor to level 1 (with all inputs set to 0,
throughout the simulation).
doi:10.1371/journal.pcbi.1003286.g006
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
11
October 2013 | Volume 9 | Issue 10 | e1003286
which covers the interplays between NFkB pro-survival pathway,
RIP1-dependent necrosis, and extrinsic/intrinsic apoptosis path-
ways.
In the Introduction, we highlighted the importance of specificity
factors in determining signal specificity of the MAPK network and
took this into consideration in the construction of the reaction
map. However, given the heterogeneity of available information
among the different MAPK cascades, we could not include all
these factors in our logical model. Nonetheless, we considered
some of them, including several feedbacks, cross-talks, phospha-
tases and input stimuli. These allowed us to focus on interesting
aspects and identify mechanisms potentially underlying the
different responses of bladder cancer cells to different growth
factor receptors (EGFR versus FGFR3).
The role of SPRY-dependent down-regulation of FGFR3
signalling seems to be determinant for the decision between
proliferative and non-proliferative response. Moreover, the model
also suggests that the presence of PI3K/AKT, but not ERK,
positively correlates with the presence of a proliferative phenotype.
Nevertheless, ERK-related mechanisms (fastness/strength of ERK
activation and activation of SPRY) seem to be determinant for
driving the switch.
Such different responses provide a striking example of how
signals transduced by largely overlapping pathways can produce
opposite effects. To explain this behaviour, we analysed the roles
of specific model circuits, which are presumably extremely
important in the phenotype choice. Our data further highlight
the contribution of cross-talks among the MAPK cascades to cell
fate decision. Other specificity factors, including scaffold proteins
and sub-cellular localisation, should also be taken into consider-
ation in the near future, as novel data on these factors will be
gathered. This will require a regular updating of our MAPK
reaction map, by including new findings related to cell fate
decision.
We interpreted the p53-independent response of the MAPK
network to FGFR3 stimulus as a sort of balance between
proliferative and non-proliferative phenotype. A decreased rate
of cell proliferation might indeed explain the less aggressive
phenotypes frequently observed in FGFR3-mutated bladder
carcinomas, in comparison with EGFR over-expression cases.
Interestingly, this behaviour can be further related with opposite
effects of FGFR3 activation in other cell types. In particular,
activating FGFR3 mutations have been associated with growth
arrest in chondrocytes, whereas they enhance proliferation and/or
transformation in several cancer types and skin disorders (e.g.
bladder cancer, multiple myeloma, seborrheic keratosis, etc.) [79].
Tentatively, proper modifications (e.g. concerning the introduction
of STAT-dependent pathways and tuning of AKT response to
growth factors [80]) may enable our MAPK model to account for
these observations.
Finally, we are currently assessing a potential proliferative role
of p38 in FGFR3-mutated bladder carcinomas (unpublished
preliminary data), which might lead to further model refinement.
To wrap up, the present study demonstrates how Boolean
modelling can recapitulate salient dynamical properties of an
extremely complex biological network. As further details are
uncovered, our logical model could be refined and eventually
translated into a more quantitative framework (e.g. using ODEs or
stochastic equations). In a first step towards more quantitative
simulations, a continuous time Boolean framework could be used
to explicitly represent time dependencies [81]. Tentatively, this
approach would allow us to recapitulate more precisely the
differential effects of transient versus sustained ERK activation
[33,37,82].
Combining the delineation of a detailed reaction map and that
of a predictive logical model, this study can serve as a basis to
develop (semi-)automatic tools to derive logical models from
reaction maps. Indeed, the manual derivation of a logical model
from a complex reaction map presents risks of misinterpretations
of either map symbols or map annotations. Errors are particularly
likely to happen when the model is not built by the author of the
map. In this respect, recent rule-based languages used to derive
more quantitative models could be used to systematically derive
predictive logical models, although potentially at the cost of
additional efforts to build reaction maps in a more rigorous fashion
[83–85].
Supporting Information
Dataset S1
MAPK reaction map. The png (map) and txt
(annotations) files were directly exported from the corresponding
CellDesigner file (Dataset S2). Map components are coloured to
emphasise relevant classes of proteins. The default protein colour
is light green, whereas the default gene colour is yellow. MAPK
cascades are coloured with different blue gradations (from light to
dark blue going down the cascade). Scaffold proteins are coloured
in darker green; phosphatases are coloured in red. Complete
graphical notations can be found at www.celldesigner.org.
(ZIP)
Dataset S2
CellDesigner file (xml format) of the MAPK reaction
map. Species and reactions are annotated and identifiers of the
corresponding sources of information (PubMed IDs) are provided.
(XML)
Dataset S3
Summary of the results of the main simulations
performed in this work. The xls file includes three sheets,
erresponding to a model reduction. For each simulation, we
report here the simulation ID (referenced in the main text and in
the model files provided in Dataset S4), the perturbations
performed (e.g. ‘‘EGFRgain; p53loss’’ indicates that EGFR was
set to 1 and p53 was set to 0 throughout the simulation), the inputs
considered and the initials states (asterisks denote all possible
combinations of initial states). The attractor types (along with the
number of corresponding states within parentheses, in the case of
cyclic attractors) are further reported, as well as the corresponding
component values in the attractor: 1 or 0 for stable values; asterisks
for oscillating values.
(XLS)
Dataset S4
GINsim model files. For each model version (the
original large model, and the three reduced versions), a file is
provided in the format (zginml) that can be opened with the
software GINsim (http://ginsim.org/beta). Simulation parameters
have been encoded to ease the reproduction of the experiments
referenced in the main text.
(ZIP)
Table S1
Selected MAPK modelling studies.
(PDF)
Table S2
Logical rules for the MAPK comprehensive model.
& = AND; | = OR; ! = NOT. More details about modelling
assumptions and references are provided in Table S4 (model
documentation).
(PDF)
Table
S3
Reduced MAPK models. We considered three
alternative reductions of the MAPK model (columns), each
preserving the input and phenotype components. Additional
components (Selected observables) were kept depending on the
simulations performed. The last row lists components that were
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
12
October 2013 | Volume 9 | Issue 10 | e1003286
conserved because they turned out to be auto-regulated at some
point during the reduction procedure. Such auto-regulations arise
from the compression of longer circuits.
(PDF)
Table S4
MAPK model documentation. Following a brief
general description, all the components of the MAPK model
(comprehensive version) are reported, along with their corre-
sponding logical rules and annotations, including modelling
hypotheses and links to the main sources of information (PubMed
and HGNC databases).
(PDF)
Text S1
Supplementary text encompassing two sections. The
first one describes how we derived the logical model from the
reaction map. The second one demonstrates how we checked that
all cyclic attractors obtained for the MAPK model reductions
indeed correspond to attractors of the original comprehensive
model.
(PDF)
Text S2
Hierarchical transition graphs associated with receptor
alterations. Model dynamics following either EGFR over-expres-
sion or FGFR3 activating mutation (with all inputs set to 0,
throughout the simulations) are depicted in two separated graphs,
which were obtained using the reduced model version red1. For
sake of simplicity, simulations were performed by using a single
initial state with all the remaining variables set to 0 (the salient
dynamics were preserved in these cases – cf. Dataset S3). The
resulting hierarchical transition graphs (see Methods) are com-
posed by different classes of nodes, emphasising strongly connected
components (blue), and linear (non circular) pathways (pink). The
attractors reached are represented at the bottom of the figures.
Attractor colours refer to the corresponding phenotypes: red for
proliferation, green for apoptosis, grey for no decision. Stables
states are denoted by rectangles, while cyclic attractors are
denoted by circles. The accompanying tables give the composition
of each node of the corresponding HTG. For instance, the node
cc1 of the HTG obtained for FGFR3 activating mutation
corresponds to a strongly connected component of the state
transition graph. The number of states belonging to it (i.e. 24), as
well as the list of these states are listed in the table (asterisks denote
all possible values for the corresponding variable).
(PDF)
Acknowledgments
We thank Emmanuel Barillot, Claudine Chaouiya, Adrien Faure´, Je´roˆme
Feret, Abibatou Mbodj and Aure´lien Naldi for many insightful discussions.
We further thank Ozgu¨r Sahin and Andrei Zinovyev for their critical
reading of earlier versions of this manuscript.
Author Contributions
Conceived and designed the experiments: LG LC FR DT. Performed the
experiments: LG. Analyzed the data: LG LC IBP FR BKP DT.
Contributed reagents/materials/analysis tools: DT. Wrote the paper: LG
LC IBP FR BKP DT.
References
1. Krishna M, Narang H (2008) The complexity of mitogen-activated protein
kinases (MAPKs) made simple. Cellular and Molecular Life Sciences 65: 3525–
3544. doi:10.1007/s00018-008-8170-7.
2. Junttila MR, Li S-P, Westermarck J (2007) Phosphatase-mediated crosstalk
between MAPK signaling pathways in the regulation of cell survival. The
FASEB Journal 22: 954–965. doi:10.1096/fj.06-7859rev.
3. Shen YH (2003) Cross-talk between JNK/SAPK and ERK/MAPK Pathways:
sustained activation of JNK blocks ERK activation by mitogenic factors. Journal
of Biological Chemistry 278: 26715–26721. doi:10.1074/jbc.M303264200.
4. Zehorai E, Yao Z, Plotnikov A, Seger R (2010) The subcellular localization of
MEK and ERK—A novel nuclear translocation signal (NTS) paves a way to the
nucleus. Molecular and Cellular Endocrinology 314: 213–220. doi:10.1016/
j.mce.2009.04.008.
5. Owens DM, Keyse SM (2007) Differential regulation of MAP kinase signalling
by dual-specificity protein phosphatases. Oncogene 26: 3203–3213.
doi:10.1038/sj.onc.1210412.
6. Morrison DK, Davis RJ (2003) Regulation of MAP kinase signaling modules by
scaffold proteins in mammals. Annual Review of Cell and Developmental
Biology 19: 91–118. doi:10.1146/annurev.cellbio.19.111401.091942.
7. Kyriakis JM, Avruch J (2001) Mammalian mitogen-activated protein kinase
signal transduction pathways activated by stress and inflammation. Physiological
reviews 81: 807–869.
8. Roux PP, Blenis J (2004) ERK and p38 MAPK-activated protein kinases: a
family of protein kinases with diverse biological functions. Microbiology and
molecular biology reviews 68: 320–344.
9. Kolch W, Calder M, Gilbert D (2005) When kinases meet mathematics: the
systems biology of MAPK signalling. FEBS Letters 579: 1891–1895.
doi:10.1016/j.febslet.2005.02.002.
10. Plotnikov A, Zehorai E, Procaccia S, Seger R (2011) The MAPK cascades:
Signaling components, nuclear roles and mechanisms of nuclear translocation.
Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 1813: 1619–
1633. doi:10.1016/j.bbamcr.2010.12.012.
11. Dhillon AS, Hagan S, Rath O, Kolch W (2007) MAP kinase signalling pathways
in cancer. Oncogene 26: 3279–3290. doi:10.1038/sj.onc.1210421.
12. Wagner EF, Nebreda A´ R (2009) Signal integration by JNK and p38 MAPK
pathways in cancer development. Nature Reviews Cancer 9: 537–549.
doi:10.1038/nrc2694.
13. Halilovic E, Solit DB (2008) Therapeutic strategies for inhibiting oncogenic
BRAF signaling. Current Opinion in Pharmacology 8: 419–426. doi:10.1016/
j.coph.2008.06.014.
14. Santarpia L, Lippman SM, El-Naggar AK (2012) Targeting the MAPK–RAS–
RAF signaling pathway in cancer therapy. Expert Opinion on Therapeutic
Targets 16: 103–119. doi:10.1517/14728222.2011.645805.
15. Zachos I, Konstantinopoulos PA, Tzortzis V, Gravas S, Karatzas A, et al. (2010)
Systemic therapy of metastatic bladder cancer in the molecular era: current
status and future promise. Expert Opinion on Investigational Drugs 19: 875–
887. doi:10.1517/13543784.2010.496450.
16. Yong H-Y, Koh M-S, Moon A (2009) The p38 MAPK inhibitors for the
treatment of inflammatory diseases and cancer. Expert Opinion on Investiga-
tional Drugs 18: 1893–1905. doi:10.1517/13543780903321490.
17. Sullivan RJ, Atkins MB (2010) Molecular targeted therapy for patients with
melanoma: the promise of MAPK pathway inhibition and beyond. Expert
Opinion
on
Investigational
Drugs
19:
1205–1216.
doi:10.1517/
13543784.2010.504709.
18. Young A, Lyons J, Miller AL, Phan VT, Alarco´n IR, et al. (2009) Chapter 1 Ras
Signaling and Therapies. Advances in Cancer Research. Elsevier, Vol. 102.
pp.
1–17.
Available:
http://linkinghub.elsevier.com/retrieve/pii/
S0065230X09020016.
19. Poulikakos PI, Solit DB (2011) Resistance to MEK inhibitors: should we co-
target upstream? Science Signalling 4: pe16.
20. Haagenson KK, Wu GS (2010) The role of MAP kinases and MAP kinase
phosphatase-1 in resistance to breast cancer treatment. Cancer and Metastasis
Reviews 29: 143–149. doi:10.1007/s10555-010-9208-5.
21. Chapman MS, Miner JN (2011) Novel mitogen-activated protein kinase kinase
inhibitors. Expert Opinion on Investigational Drugs 20: 209–220. doi:10.1517/
13543784.2011.548803.
22. Cakir M, Grossman AB (2009) Targeting MAPK (Ras/ERK) and PI3K/Akt
pathways in pituitary tumorigenesis. Expert Opinion on Therapeutic Targets 13:
1121–1134. doi:10.1517/14728220903170675.
23. Montagut C, Settleman J (2009) Targeting the RAF–MEK–ERK pathway in
cancer therapy. Cancer Letters 283: 125–134. doi:10.1016/j.canlet.
2009.01.022.
24. Inamdar GS, Madhunapantula SV, Robertson GP (2010) Targeting the MAPK
pathway in melanoma: Why some approaches succeed and other fail.
Biochemical Pharmacology 80: 624–637. doi:10.1016/j.bcp.2010.04.029.
25. Orton RJ, Sturm OE, Vyshemirsky V, Calder M, Gilbert DR, et al. (2005)
Computational modelling of the receptor-tyrosine-kinase-activated MAPK
pathway. Biochemical Journal 392: 249. doi:10.1042/BJ20050908.
26. Huang CY, Ferrell JE (1996) Ultrasensitivity in the mitogen-activated protein kinase
cascade. Proceedings of the National Academy of Sciences 93: 10078–10083.
27. Kholodenko BN (2001) Negative feedback and ultrasensitivity can bring about
oscillations in the mitogen-activated protein kinase cascades. European Journal
of Biochemistry 267: 1583–1588.
28. Levchenko A, Bruck J, Sternberg PW (2000) Scaffold proteins may biphasically
affect the levels of mitogen-activated protein kinase signaling and reduce its
threshold properties. Proceedings of the National Academy of Sciences 97:
5818–5823.
29. Markevich NI (2004) Signaling switches and bistability arising from multisite
phosphorylation in protein kinase cascades. The Journal of Cell Biology 164:
353–359. doi:10.1083/jcb.200308060.
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
13
October 2013 | Volume 9 | Issue 10 | e1003286
30. Legewie S, Schoeberl B, Blu¨thgen N, Herzel H (2007) Competing Docking
Interactions can Bring About Bistability in the MAPK Cascade. Biophysical
Journal 93: 2279–2288. doi:10.1529/biophysj.107.109132.
31. Smolen P, Baxter DA, Byrne JH (2007) Bistable MAP kinase activity: a plausible
mechanism contributing to maintenance of late long-term potentiation. AJP:
Cell Physiology 294: C503–C515. doi:10.1152/ajpcell.00447.2007.
32. Sturm OE, Orton R, Grindlay J, Birtwistle M, Vyshemirsky V, et al. (2010) The
mammalian MAPK/ERK pathway exhibits properties of a negative feedback
amplifier. Science Signalling 3: ra90.
33. Brightman FA, Fell DA (2000) Differential feedback regulation of the MAPK
cascade underlies the quantitative differences in EGF and NGF signalling in
PC12 cells. FEBS letters 482: 169–174.
34. Schoeberl B, Eichler-Jonsson C, Gilles ED, Muller G (2002) Computational
modeling of the dynamics of the MAP kinase cascade activated by surface and
internalized EGF receptors. Nature biotechnology 20: 370–375.
35. Hatakeyama M, Kimura S, Naka T, Kawasaki T, Yumoto N, et al. (2003) A
computational model on the modulation of mitogen-activated protein kinase
(MAPK) and Akt pathways in heregulin-induced ErbB signalling. Biochemical
Journal 373: 451.
36. Hornberg JJ, Binder B, Bruggeman FJ, Schoeberl B, Heinrich R, et al. (2005)
Control of MAPK signalling: from complexity to what really matters. Oncogene
24: 5533–5542.
37. Sasagawa S, Ozaki Y, Fujita K, Kuroda S (2005) Prediction and validation of
the distinct dynamics of transient and sustained ERK activation. Nature Cell
Biology 7: 365–373. doi:10.1038/ncb1233.
38. Orton RJ, Adriaens ME, Gormand A, Sturm OE, Kolch W, et al. (2009)
Computational modelling of cancerous mutations in the EGFR/ERK signalling
pathway. BMC Systems Biology 3: 100. doi:10.1186/1752-0509-3-100.
39. Chen WW, Schoeberl B, Jasper PJ, Niepel M, Nielsen UB, et al. (2009) Input–
output behavior of ErbB signaling pathways as revealed by a mass action model
trained against dynamic data. Molecular Systems Biology 5: 239. Available:
http://www.nature.com/doifinder/10.1038/msb.2008.74.
40. Bagheri N, Shiina M, Lauffenburger DA, Korn WM (2011) A Dynamical
Systems Model for Combinatorial Cancer Therapy Enhances Oncolytic
Adenovirus Efficacy by MEK-Inhibition. PLoS Computational Biology 7:
e1001085. doi:10.1371/journal.pcbi.1001085.
41. Handorf T, Klipp E (2011) Modeling mechanistic biological networks: An
advanced Boolean approach. Bioinformatics 28: 557–563. doi:10.1093/
bioinformatics/btr697.
42. Finch AR, Caunt CJ, Perrett RM, Tsaneva-Atanasova K, McArdle CA (2012)
Dual specificity phosphatases 10 and 16 are positive regulators of EGF-
stimulated ERK activity: Indirect regulation of ERK signals by JNK/p38
selective MAPK phosphatases. Cellular Signalling 24: 1002–1011. doi:10.1016/
j.cellsig.2011.12.021.
43. Samaga R, Saez-Rodriguez J, Alexopoulos LG, Sorger PK, Klamt S (2009) The
Logic of EGFR/ErbB Signaling: Theoretical Properties and Analysis of High-
Throughput Data. PLoS Computational Biology 5: e1000438. doi:10.1371/
journal.pcbi.1000438.
44. Oda K, Matsuoka Y, Funahashi A, Kitano H (2005) A comprehensive pathway
map of epidermal growth factor receptor signaling. Molecular Systems Biology
1: E1–E17. doi:10.1038/msb4100014.
45. Poltz R, Naumann M (2012) Dynamics of p53 and NF-kappaB regulation in
response to DNA damage and identification of target proteins suitable for
therapeutic intervention. BMC Systems Biology 6: 125.
46. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100: 57–70.
47. Ho JR, Chapeaublanc E, Kirkwood L, Nicolle R, Benhamou S, et al. (2012)
Deregulation of Rab and Rab Effector Genes in Bladder Cancer. PLoS One 7:
e39469. doi:10.1371/journal.pone.0039469.
48. Billerey C, Chopin D, Aubriot-Lorton MH, Ricol D, de Medina SGD, et al.
(2001) Frequent FGFR3 mutations in papillary non-invasive bladder (pTa)
tumors. The American journal of pathology 158: 1955.
49. Williams SV, Hurst CD, Knowles MA (2012) Oncogenic FGFR3 gene fusions in
bladder cancer. Human Molecular Genetics 22(4):795–803. Available: http://
www.hmg.oxfordjournals.org/cgi/doi/10.1093/hmg/dds486.
50. Mitra AP, Cote RJ (2009) Molecular Pathogenesis and Diagnostics of Bladder
Cancer. Annual Review of Pathology: Mechanisms of Disease 4: 251–285.
doi:10.1146/annurev.pathol.4.110807.092230.
51. Mitra AP, Datar RH, Cote RJ (2006) Molecular Pathways in Invasive Bladder
Cancer: New Insights Into Mechanisms, Progression, and Target Identification.
Journal of Clinical Oncology 24: 5552–5564. doi:10.1200/JCO.2006.08.2073.
52. Zindy F, Eischen CM, Randle DH, Kamijo T, Cleveland JL, et al. (1998) Myc
signaling via the ARF tumor suppressor regulates p53-dependent apoptosis and
immortalization. Genes Dev 12: 2424–2433.
53. Funahashi A, Morohashi M, Kitano H, Tanimura N (2003) CellDesigner: a
process diagram editor for gene-regulatory and biochemical networks.
BIOSILICO 1: 159–162. doi:10.1016/S1478-5382(03)02370-9.
54. Kitano H, Funahashi A, Matsuoka Y, Oda K (2005) Using process diagrams for
the graphical representation of biological networks. Nature Biotechnology 23:
961–966. doi:10.1038/nbt1111.
55. Funahashi A, Matsuoka Y, Jouraku A, Morohashi M, Kikuchi N, et al. (2008)
CellDesigner 3.5: A Versatile Modeling Tool for Biochemical Networks.
Proceedings of the IEEE 96: 1254–1265. doi:10.1109/JPROC.2008.925458.
56. Naldi A, Berenguier D, Faure´ A, Lopez F, Thieffry D, et al. (2009) Logical
modelling of regulatory networks with GINsim 2.3. Biosystems 97: 134–139.
doi:10.1016/j.biosystems.2009.04.008.
57. Chaouiya C, Naldi A, Thieffry D (2012) Logical Modelling of Gene Regulatory
Networks with GINsim. Methods in Molecular Biology 804: 463–479. Available:
http://www.springerlink.com/index/10.1007/978-1-61779-361-5_23.
58. Thomas R (1991) Regulatory networks seen as asynchronous automata: A
logical description. Journal of Theoretical Biology 153: 1–23. doi:10.1016/
S0022-5193(05)80350-9.
59. Thomas R, Thieffry D, Kaufman M (1995) Dynamical behaviour of biological
regulatory networks—I. Biological role of feedback loops and practical use of the
concept of the loop-characteristic state. Bulletin of Mathematical Biology 57:
247–276.
60. Chaouiya C, Remy E, Mosse´ B, Thieffry D (2003) Qualitative analysis of
regulatory graphs: a computational tool based on a discrete formal framework.
Positive Systems: 830–832.
61. Faure A, Naldi A, Chaouiya C, Thieffry D (2006) Dynamical analysis of a
generic Boolean model for the control of the mammalian cell cycle.
Bioinformatics 22: e124–e131. doi:10.1093/bioinformatics/btl210.
62. Naldi A, Thieffry D, Chaouiya C (2007) Decision diagrams for the
representation and analysis of logical models of genetic networks. Computa-
tional Methods in Systems Biology. pp. 233–247. Available: http://www.
springerlink.com/index/ww3402468t532578.pdf.
63. Naldi A, Remy E, Thieffry D, Chaouiya C (2011) Dynamically consistent
reduction of logical regulatory graphs. Theoretical Computer Science 412:
2207–2218. doi:10.1016/j.tcs.2010.10.021.
64. Be´renguier D, Chaouiya C, Monteiro PT, Naldi A, Remy E, et al. (2013)
Dynamical modeling and analysis of large cellular regulatory networks. Chaos
23: 025114. doi: 10.1063/1.4809783.
65. Thomas R (1981) On the relation between the logical structure of systems and
their ability to generate multiple steady states or sustained oscillations. Springer
Series in Synergetics 9: 180–193.
66. Remy E´, Ruet P, Thieffry D (2008) Graphic requirements for multistability and
attractive cycles in a Boolean dynamical framework. Advances in Applied
Mathematics 41: 335–350. doi:10.1016/j.aam.2007.11.003.
67. Thieffry D (2007) Dynamical roles of biological regulatory circuits. Briefings in
Bioinformatics 8: 220–225. doi:10.1093/bib/bbm028.
68. Weinberg RA (2007) The Biology of cancer. New York: Garland Science.
69. Takekawa M, Kubota Y, Nakamura T, Ichikawa K (2011) Regulation of stress-
activated MAP kinase pathways during cell fate decisions. Nagoya journal of
medical science 73: 1–14.
70. Heldin C-H, Landstro¨m M, Moustakas A (2009) Mechanism of TGF-b signaling
to growth arrest, apoptosis, and epithelial–mesenchymal transition. Current
Opinion in Cell Biology 21: 166–176. doi:10.1016/j.ceb.2009.01.021.
71. Tanaka M, Koul D, Davies MA, Liebert M, Steck PA, et al. (2000) MMAC1/
PTEN inhibits cell growth and induces chemosensitivity to doxorubicin in
human bladder cancer cells. Oncogene 19: 5406–5412. doi:10.1038/
sj.onc.1203918.
72. Sears RC (2002) Signaling Networks That Link Cell Proliferation and Cell Fate.
Journal of Biological Chemistry 277: 11617–11620. doi:10.1074/
jbc.R100063200.
73. Schulz WA (2006) Understanding urothelial carcinoma through cancer
pathways. International Journal of Cancer 119: 1513–1518. doi:10.1002/
ijc.21852.
74. Cho JY (2004) Defective lysosomal targeting of activated fibroblast growth factor
receptor 3 in achondroplasia. Proceedings of the National Academy of Sciences
101: 609–614. doi:10.1073/pnas.2237184100.
75. Schlessinger J (2004) Common and Distinct Elements in Cellular Signaling via
EGF and FGF Receptors. Science 306: 1506–1507. doi:10.1126/science.
1105396.
76. Cochet C, Gill GN, Meisenhelder J, Cooper JA, Hunter T (1984) C-kinase
phosphorylates the epidermal growth factor receptor and reduces its epidermal
growth factor-stimulated tyrosine protein kinase activity. Journal of Biological
Chemistry 259: 2553–2558.
77. Li L, Chen SF, Liu Y (2009) MAP kinase phosphatase-1, a critical negative
regulator of the innate immune response. International journal of clinical and
experimental medicine 2: 48.
78. Calzone L, Tournier L, Fourquet S, Thieffry D, Zhivotovsky B, et al. (2010)
Mathematical Modelling of Cell-Fate Decision in Response to Death Receptor
Engagement. PLoS Computational Biology 6: e1000702. doi:10.1371/journal.
pcbi.1000702.
79. Foldynova-Trantirkova S, Wilcox WR, Krejci P (2012) Sixteen years and
counting: The current understanding of fibroblast growth factor receptor 3
(FGFR3) signaling in skeletal dysplasias. Human Mutation 33: 29–41.
doi:10.1002/humu.21636.
80. Priore R, Dailey L, Basilico C (2006) Downregulation of Akt activity contributes
to the growth arrest induced by FGF in chondrocytes. J Cell Physiol 207: 800–
808. doi:10.1002/jcp.20620.
81. Stoll G, Viara E, Barillot E, Calzone L (2012) Continuous time Boolean
modeling for biological signaling: application of Gillespie algorithm. BMC
Systems Biology 6: 116. doi:10.1186/1752-0509-6-116.
82. Roovers K, Assoian RK (2000) Integrating the MAP kinase signal into the G 1
phase cell cycle machinery. Bioessays 22: 818–826.
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
14
October 2013 | Volume 9 | Issue 10 | e1003286
83. Blinov ML, Faeder JR, Goldstein B, Hlavacek WS (2006) A network model of
early events in epidermal growth factor receptor signaling that accounts for
combinatorial complexity. Biosystems 83: 136–151. doi:10.1016/j.biosystems.
2005.06.014.
84. Feret J, Danos V, Krivine J, Harmer R, Fontana W (2009) Internal coarse-
graining of molecular systems.Proceedings of the National Academy of Sciences
of the United States of America 106: 6453–6458. doi: 10.1073/
pnas.0809908106.
85. Fages F, Soliman S (2008) From reaction models to influence graphs and back: a
theorem. Formal Methods in Systems Biology 5054: 90–102.
86. Sharp S, Workman P (2006) Inhibitors of the HSP90 Molecular Chaperone:
Current Status. Advances in Cancer Research. Elsevier, Vol. 95. pp. 323–348.
Available: http://linkinghub.elsevier.com/retrieve/pii/S0065230X0695009X.
87. Stein JP, Ginsberg DA, Grossfeld GD, Chatterjee SJ, Esrig D, et al. (1998) Effect
of p21WAF1/CIP1 expression on tumor progression in bladder cancer. Journal
of the National Cancer Institute 90: 1072–1079.
88. Kennedy NJ, Davis RJ (2003) Role of JNK in tumor development. Cell Cycle 2:
199–201.
89. Schramek H (2002) MAP kinases: from intracellular signals to physiology and
disease. Physiology 17: 62–67.
Modelling of MAPK Influence on Cell Fate Decision
PLOS Computational Biology | www.ploscompbiol.org
15
October 2013 | Volume 9 | Issue 10 | e1003286
|
24250280
|
PDK1 = ( PI3K )
FRS2 = ( ( ( FGFR3 ) AND NOT ( SPRY ) ) AND NOT ( GRB2 ) )
MAX = ( p38 )
GAB1 = ( GRB2 ) OR ( PI3K )
PI3K = ( GAB1 ) OR ( RAS AND ( ( ( SOS ) ) ) )
AKT = ( ( PDK1 ) AND NOT ( PTEN ) )
p70 = ( PDK1 AND ( ( ( ERK ) ) ) )
ERK = ( MEK1_2 )
SOS = ( ( GRB2 ) AND NOT ( RSK ) )
JNK = ( TAK1 AND ( ( ( MAP3K1_3 ) ) ) ) OR ( MAP3K1_3 AND ( ( ( MTK1 ) ) ) ) OR ( MTK1 AND ( ( ( NOT DUSP1 ) ) ) ) OR ( TAOK AND ( ( ( NOT DUSP1 ) ) ) )
EGFR = ( ( ( SPRY ) AND NOT ( GRB2 ) ) AND NOT ( PKC ) ) OR ( ( ( EGFR_stimulus ) AND NOT ( GRB2 ) ) AND NOT ( PKC ) )
p53 = ( p38 AND ( ( ( NOT MDM2 ) ) ) ) OR ( ATM AND ( ( ( p38 ) ) ) )
RAF = ( ( ( PKC ) AND NOT ( AKT ) ) AND NOT ( ERK ) ) OR ( ( ( RAS ) AND NOT ( AKT ) ) AND NOT ( ERK ) )
RAS = ( SOS ) OR ( PLCG )
GRB2 = ( EGFR ) OR ( FRS2 ) OR ( TGFBR )
MSK = ( p38 ) OR ( ERK )
SPRY = ( ERK )
MYC = ( MSK AND ( ( ( MAX ) ) ) )
RSK = ( ERK )
FOXO3 = ( ( JNK ) AND NOT ( AKT ) )
MEK1_2 = ( ( ( RAF ) AND NOT ( PPP2CA ) ) AND NOT ( AP1 ) ) OR ( ( ( MAP3K1_3 ) AND NOT ( PPP2CA ) ) AND NOT ( AP1 ) )
PPP2CA = ( p38 )
DUSP1 = ( CREB )
PLCG = ( EGFR ) OR ( FGFR3 )
FOS = ( ERK AND ( ( ( RSK ) AND ( ( ( ELK1 OR CREB ) ) ) ) ) )
TGFBR = ( TGFBR_stimulus )
ATM = ( DNA_damage )
JUN = ( JNK )
Proliferation = ( ( p70 AND ( ( ( MYC ) ) ) ) AND NOT ( p21 ) )
GADD45 = ( SMAD ) OR ( p53 )
p21 = ( ( p53 ) AND NOT ( AKT ) )
FGFR3 = ( ( ( FGFR3_stimulus ) AND NOT ( PKC ) ) AND NOT ( GRB2 ) )
MAP3K1_3 = ( RAS )
TAOK = ( ATM )
AP1 = ( JUN AND ( ( ( ATF2 OR FOS ) ) ) )
ELK1 = ( p38 ) OR ( JNK ) OR ( ERK )
PKC = ( PLCG )
BCL2 = ( CREB AND ( ( ( AKT ) ) ) )
PTEN = ( p53 )
p38 = ( TAK1 AND ( ( ( NOT DUSP1 ) ) ) ) OR ( MAP3K1_3 AND ( ( ( NOT DUSP1 ) ) ) ) OR ( MTK1 AND ( ( ( NOT DUSP1 ) ) ) ) OR ( TAOK AND ( ( ( MTK1 ) ) ) )
Apoptosis = ( ( ( FOXO3 AND ( ( ( p53 ) ) ) ) AND NOT ( ERK ) ) AND NOT ( BCL2 ) )
Growth_Arrest = ( p21 )
MDM2 = ( ( AKT ) AND NOT ( p14 ) ) OR ( ( p53 ) AND NOT ( p14 ) )
CREB = ( MSK )
TAK1 = ( TGFBR )
MTK1 = ( GADD45 )
ATF2 = ( p38 ) OR ( JNK )
p14 = ( MYC )
SMAD = ( TGFBR )
|
Boolean ErbB network reconstructions and
perturbation simulations reveal individual drug
response in different breast cancer cell lines
von der Heyde et al.
von der Heyde et al. BMC Systems Biology 2014, 8:75
http://www.biomedcentral.com/1752-0509/8/75
von der Heyde et al. BMC Systems Biology 2014, 8:75
http://www.biomedcentral.com/1752-0509/8/75
RESEARCH ARTICLE
Open Access
Boolean ErbB network reconstructions and
perturbation simulations reveal individual drug
response in different breast cancer cell lines
Silvia von der Heyde1, Christian Bender2, Frauke Henjes3, Johanna Sonntag4, Ulrike Korf4
and Tim Beißbarth1*
Abstract
Background: Despite promising progress in targeted breast cancer therapy, drug resistance remains challenging.
The monoclonal antibody drugs trastuzumab and pertuzumab as well as the small molecule inhibitor erlotinib were
designed to prevent ErbB-2 and ErbB-1 receptor induced deregulated protein signalling, contributing to tumour
progression. The oncogenic potential of ErbB receptors unfolds in case of overexpression or mutations. Dimerisation
with other receptors allows to bypass pathway blockades. Our intention is to reconstruct the ErbB network to reveal
resistance mechanisms. We used longitudinal proteomic data of ErbB receptors and downstream targets in the ErbB-2
amplified breast cancer cell lines BT474, SKBR3 and HCC1954 treated with erlotinib, trastuzumab or pertuzumab,
alone or combined, up to 60 minutes and 30 hours, respectively. In a Boolean modelling approach, signalling
networks were reconstructed based on these data in a cell line and time course specific manner, including prior
literature knowledge. Finally, we simulated network response to inhibitor combinations to detect signalling nodes
reflecting growth inhibition.
Results: The networks pointed to cell line specific activation patterns of the MAPK and PI3K pathway. In BT474, the
PI3K signal route was favoured, while in SKBR3, novel edges highlighted MAPK signalling. In HCC1954, the inferred
edges stimulated both pathways. For example, we uncovered feedback loops amplifying PI3K signalling, in line with
the known trastuzumab resistance of this cell line. In the perturbation simulations on the short-term networks, we
analysed ERK1/2, AKT and p70S6K. The results indicated a pathway specific drug response, driven by the type of
growth factor stimulus. HCC1954 revealed an edgetic type of PIK3CA-mutation, contributing to trastuzumab inefficacy.
Drug impact on the AKT and ERK1/2 signalling axes is mirrored by effects on RB and RPS6, relating to phenotypic
events like cell growth or proliferation. Therefore, we additionally analysed RB and RPS6 in the long-term networks.
Conclusions: We derived protein interaction models for three breast cancer cell lines. Changes compared to the
common reference network hint towards individual characteristics and potential drug resistance mechanisms.
Simulation of perturbations were consistent with the experimental data, confirming our combined reverse and
forward engineering approach as valuable for drug discovery and personalised medicine.
Keywords: ErbB, RPPA, Network reconstruction, Boolean model, Breast cancer cell line, Drug resistance
*Correspondence: tim.beissbarth@ams.med.uni-goettingen.de
1Statistical Bioinformatics, Department of Medical Statistics, University Medical
Center Göttingen, Humboldtallee 32, 37073 Göttingen, Germany
Full list of author information is available at the end of the article
© 2014 von der Heyde et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the
Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use,
distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public
Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this
article, unless otherwise stated.
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 2 of 21
http://www.biomedcentral.com/1752-0509/8/75
Background
Longitudinal time course data are the basis for modelling
signalling networks in a holistic systems biology approach
in order to uncover mechanisms of signal transduction
dynamics [1,2]. Network models provide novel insight
[3,4] and allow us to perform efficiently simulations to
predict systems behaviour or evaluate certain hypotheses
[5]. Furthermore, combining perturbation experiments
with the measurements of system dynamics seems to
be even more efficient than time series data on their
own [6-8]. Knock-outs or stimuli as directed perturba-
tions support the systematic identification of regulatory
relationships.
Quantitative models, based on differential equations,
require explicit knowledge on the kinetics of the sys-
tem of interest [9-12]. In contrast, the qualitative Boolean
abstraction considers the components’ states as binary
variables, being either active (1) or passive (0), but nev-
ertheless encompasses the essential functionality [13,14].
Wang et al. stressed, that Boolean models have already
been successfully applied in reverse engineering of pro-
teomic signalling networks, and their reduced complexity
is considered to be especially advantageous for large-
scale systems [15]. To avoid the drawbacks of purely data-
or literature-driven algorithms regarding completeness,
generalisation or interpretability, combined approaches
become more and more prominent in the area of net-
work reconstruction [6,16,17]. Some reverse engineering
approaches, like ddepn [6] or CellNOptR [18], ideally join
perturbed time course input data and literature prior
knowledge in network reconstruction, while preserving
the simplicity of Boolean logic at the same time. Forward
engineering methods allow subsequent analysis of the sta-
ble states of the reconstructed system. Hence, this may
allow to deduce possible long-term behaviour of compo-
nents activity under perturbations. Such approaches are
integrated and freely available in the open source Python
software package BooleanNet [19] or in the R [20] pack-
age BoolNet [21], for example. As reviewed by Samaga
and Klampt [22], several software tools can be applied for
the dynamic modeling of logical signal transduction net-
works. Among others, they exemplarily mentioned GIN-
sim [23], SQUAD [24], BooleanNet [19], ChemChains [25],
Odefy [26], and BoolNet [21].
Here we focus on protein signalling networks in breast
cancer, representing the most common cancer type
among women [27]. Breast cancer, as a heterogeneous
disease, can be divided into subgroups, which differ in
cellular properties as well as in prognosis. This requires
individual therapy approaches, which are in the focus of
current research and have partially already been realised.
Here we are interested in the ‘HER2-positive’ subtype
of breast cancer, overexpressing the human epidermal
growth factor receptor 2 (HER2, also termed ErbB-2).
ErbB-2 is a receptor tyrosine kinase (RTK) and mem-
ber of the epidermal growth factor (EGF) receptor family,
consisting of three further RTKs, namely ErbB-1, ErbB-3
and ErbB-4. These receptors cooperatively function as
homo- or heterodimers after activation via growth fac-
tors like EGF for ErbB-1 or heregulin (HRG) for ErbB-3
[28]. This initialises signalling cascades, pathologically
contributing to tumourigenesis and tumour progression.
Interestingly, different dimer formations induce different
signalling pathways, like PI3K and MAPK, also with dif-
fering signalling strengths [29]. The role of the orphan
receptor ErbB-2 in dysregulation of the ErbB network
is of major interest, due to its overexpression in 10-
20% of breast tumours, diagnosed as HER2-positive.
Furthermore, its role as favoured dimerisation part-
ner independent on ligand-activation implies oncogenic
potential [30-32]. The therapeutic antibodies trastuzumab
and pertuzumab have especially been designed to target
ErbB-2 [33].
However,
frequently
occurring
therapy
resistance
reduces the efficiency of targeted therapeutics [34-36].
This resistance is often associated with deregulated path-
way activity [37,38] or bypasses via other RTKs, especially
ErbB family members [39]. Mainly ErbB-1 expression has
been anticipated as molecular cause to overcome impact
of ErbB-2 targeting drugs. Small-molecule inhibitors such
as erlotinib are already in use against non-small cell lung
cancer [40] and pancreatic cancer [41].
Here we aim as a first step at the identification of
individual drug response patterns and insights into drug
resistance in HER2-positive breast cancer. ErbB-2 ampli-
fied cell lines were therefore subjected to short- and
long-term drug treatment with erlotinib, pertuzumab and
trastuzumab, alone or in combinations. Samples were
analysed by reverse-phase protein arrays (RPPA) [42].
We were interested in synergistic benefits of combining
erlotinib, pertuzumab or trastuzumab in ErbB-1 express-
ing, ErbB-2 amplified tumours with differing resistance
phenotypes. Therefore three representative breast cancer
cell lines were selected as model systems, namely BT474,
SKBR3 and HCC1954, of which the latter is known to be
trastuzumab resistant due to a PIK3CA mutation, while
BT474 exhibits wild type behaviour [43]. The SKBR3 cell
line is supposed to be pertuzumab resistant [44].
ErbB dimers predominantly activate the MAPK and
PI3K pathway [29]. Therefore, we concentrated on the
involved key regulators in fast downstream signalling.
Among those were ERK1/2 and AKT, and also p70S6K,
which is upstream influenced by both of the signalling
axes. Phosphorylation of RPS6 and RB was used as long-
term indicator for proliferation, cell cycle or tumour
progression [28]. Prior literature knowledge on ErbB sig-
nalling was used as input for protein network reconstruc-
tion per cell line via ddepn. Beyond that, we inferred
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 3 of 21
http://www.biomedcentral.com/1752-0509/8/75
combined therapies that target ErbB family members, cus-
tomised to the topology of the different subtypes. BoolNet
was applied to compute stable cycles of protein activ-
ity states, so-called attractors, incorporating all possible
treatment combinations. This way, optimal drug treat-
ment to deactivate oncogenic proteins was identified.
Methods
Data
Protein abundance and phosphorylation measurements in
BT474, SKBR3 and HCC1954 cells were carried out as
described by Henjes et al. [28]. In principle, the RPPA pro-
tein array technology works as follows. Minimal amounts
(1 nl volume) of cell lysate are spotted along with a serial
dilution of control samples on nitrocellulose-coated glass
slides using a printing robot (Aushon 2470 arrayer). Sam-
ples are organised as ordered subarrays so that they are
addressable during the data analysis procedure, and a sin-
gle slide can accommodate one or more subarrays. Each
subarray is analysed using a highly specific detection anti-
body to measure the abundance of a certain protein or its
phosphorylation rate. For each spot, the ratio of bound
detection antibody is visualised using secondary antibod-
ies labelled with near infrared (NIR) fluorescent dyes.
Slides are scanned using the Odyssey scanner (LiCor Bio-
sciences). Spot intensities are determined using a microar-
ray image analysis software (GenePix).
Apart from the quantitative character, another advan-
tage of the technology is the handling of large sample sets
which protein abundance can be detected simultaneously
in a high throughput fashion. 20-200 identical slides can
be produced in parallel in a single print run.
In order to normalise the data spot-wise for deviant total
protein concentrations due to spotting variance, staining
with Fast Green FCF dye was employed [42]. There-
fore, one slide was stained with the dye to determine the
total protein content of each lysate spot and correspond-
ing signal intensity correction factors. The spots on the
remaining slides were divided by these correction factors
and afterwards multiplied by the median value to scale the
data back to the native range.
The RPPA data used here include data presented in
Henjes et al. [28]. Additionally, further targets have been
measured and were used for network reconstruction. The
complete data set has been submitted to the Gene Expres-
sion Omnibus (GEO) with accession number GSE50109.
Short-term measurements
In the short-term measurements, trastuzumab, per-
tuzumab and erlotinib were added to the cells in starva-
tion medium one hour before stimulation with the growth
factors EGF and HRG. All possible 24 combinations of
drugs and stimuli were measured. Application of the stim-
uli was defined as time point zero in the measurements.
The growth factors were chosen to activate explicitly the
MAPK and PI3K pathway. Lysate preparation was per-
formed at ten time points, namely after 0, 4, 8, 12, 16,
20, 30, 40, 50 and 60 minutes. The drug treatment exper-
iments comprised three biological replicates, whereas the
inhibitor-free experiments incorporated five biological
replicates. The experiments for the SKBR3 cell line com-
prised only two biological replicates of HRG stimulated
cells under the triple drug combination. Each biological
replicate was spotted in triplicate on the RPPA slides.
To obtain short-term signal intensities, eleven antibodies
for specific phosphorylation sites were selected according
to quality checks, including inspection of corresponding
dilution series and comparison to signals arising from sec-
ondary antibodies only. The chosen target proteins and
respective antibodies are listed in Additional file 1.
Long-term measurements
For long-term measurements, no explicit ligand stimu-
lation was performed. Instead, cells were incubated in
full growth medium for 24 hours prior to adding the
three mentioned therapeutics in double combinations or
as triplet. Single drug treatment was just conducted with
erlotinib. Full growth medium was used to avoid con-
founding effects of nutrient deficiency. Protein abundance
was also quantified without any drug application. The
measuring points included 0, 1, 2, 4, 6, 8, 12, 18, 24 and 30
hours with three biological and technical replicates each.
At time point 18, only two biological replicates were avail-
able. Additional file 1 displays the 21 targets of interest for
long-term signalling.
Statistical inference of drug effects
To determine, whether a specific drug treatment revealed
an inhibiting effect on the signal intensities of the proteins,
we applied the following method. Firstly, for each protein
and (combinatorial) drug treatment we linearly modelled
the signal intensities as depending on the factors time and
group, i.e. no drug treatment versus drug treatment. If the
interaction of both factors showed a significant (p-value <
0.05) influence on the signal intensity, we further applied
a Wilcoxon rank sum test for the measurements at time
point 60 minutes for the short-term data, or at time point
30 hours for the long-term data. Thereby, we tested for
significantly (p-value < 0.05) smaller intensity values in
the drug treated group. The drug treatments with a sig-
nificant test result were considered as efficient inhibitors.
The therapeutic (combination) with the smallest p-value
was defined as the optimal one.
Literature prior knowledge
We manually determined two reference networks, i.e. one
for each time course, as initial joint hypotheses for all
of the three breast cancer cell lines. Because emphasis
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 4 of 21
http://www.biomedcentral.com/1752-0509/8/75
was put on phosphoproteomic signalling, this was mainly
based on PhosphoSitePlus® [45]. Several publications con-
firm these assumptions, as depicted in Additional file 2.
Network reconstruction
For Boolean network reconstruction, we chose the
method of dynamic deterministic effects propagation net-
works (DDEPN) [6]. This method was particularly tailored
to perturbed longitudinal protein phosphorylation data.
It is based on the DEPN approach [46], which stands for
deterministic effects propagation networks. The deter-
minism is related to the way of perturbation effect prop-
agation in the networks from parent to child nodes,
implying transitively closed graphs. The dynamic version
of Bender et al. [6,47] differs with respect to the integra-
tion of perturbed time course measurements. While the
DEPN approach requires many perturbations, like knock-
downs, but only few time points, which are regarded as
independent measurements, ddepn is designed for longer
time series without the necessity of many or all network
nodes being perturbed. The latter situation, i.e. few per-
turbations by drug interventions, reflected the design of
the RPPA experiments under consideration here, hence
leading to the application of ddepn. Most network recon-
struction algorithms have been designed for gene expres-
sion data from microarray measurements [7], which differ
from (phospho-)protein data regarding the amount of
involved network nodes. Many current methods are tai-
lored to the inference of gene regulatory networks based
on static measurements at one time point, reflecting the
steady state of the system under consideration [48]. The
longitudinal time course data used here require a suitable
method, as provided by ddepn. The method of Bender
et al. was shown to outperform two dynamical Bayesian
network approaches, and to be capable of inferring known
signalling cascades in the ErbB pathway [47]. A further
advantage was the public availability of ddepn as an R [20]
package.
The reconstruction procedure is depicted in Additional
file
3, and the core elements are described according
to [6,47] in the following. The protein interaction net-
works are modelled as directed, possibly cyclic, graphs,
with nodes V = {vi : i ∈1, . . . , N} representing proteins
and edges representing interactions. Also the external per-
turbations, i.e. the drugs and growth factors in our case,
are modelled as nodes. The edge types can be either acti-
vating or inhibiting, denoted by 1 and -1, respectively,
in the adjacency matrix = V × V →{0, 1, −1} of
the network. An entry of zero indicates no edge between
two nodes. So each edge incorporates a pair of nodes
φij : i, j ∈1, . . . , N
. The measurement data, which form
the basis for the reconstruction, are stored in a matrix
D = {ditr : i ∈1, . . . , N, t ∈1, . . . , T, r ∈1, . . . , R}, consid-
ering T time points and R replicates.
For the inference of a network structure, optimally
fitting to the data, we applied the stochastic Markov
Chain Monte Carlo (MCMC) approach of ddepn, called
inhibMCMC, in which the space of possible networks
is sampled, based on posterior probabilities. It extends
a Metropolis-Hastings type of MCMC sampler by the
capability of sampling two edge types directly, i.e. acti-
vation and inhibition. The posterior distribution of a
network given the data D, is defined as P(|D) =
P(D|)P()
P(D)
∝P(D|)P(), with P() as the prior prob-
ability distribution and P(D|) as the likelihood of the
data given the network. The latter is defined in [47]
as p(D|)
=
p(D| ˆ∗, ˆ)
=
T
t=1
N
i=1
R
r=1
p(ditr| ˆθi ˆγ ∗
itr),
where ∗=
γ ∗
itr : i ∈1, . . . , N, t ∈1, . . . , T, r ∈1, . . . , R
denotes the optimized system state matrix, containing
active and passive states per protein and time point. It is
estimated in the following way. Assuming that the pro-
teins can be either active (1) or inactive (0), signalling
dynamics are modelled by Boolean signal propagation
for a given network. All nodes, except the permanently
active perturbations, are therefore initialised with inactive
states. The transition rule is that children nodes get acti-
vated if at least one activating parent node is active and
all inhibiting ones are inactive. In this way, all reachable
system states are computed and stored in a matrix =
{γik ∈{0, 1} : i ∈1, . . . , N, k ∈1, . . . , M}, holding column-
wise the activation states of all proteins at transition step
k. The amount of transitions is limited by 0 < M ≤2N.
This state matrix has to be optimized, as it is not related
to the measured time points yet. The true unknown state
sequence over time is represented by ∗, which is esti-
mated by a hidden Markov model (HMM). The resulting
ˆ∗indicates whether a data point ditr has an underlying
active (1) or passive (0) normal distribution
ditr ∼
N (μi0, σi0),
if ˆγ ∗
itr = 0
N (μi1, σi1),
if ˆγ ∗
itr = 1.
The distribution parameters are for each protein esti-
mated as empirical mean and standard deviation of all
measurements for the considered protein in the cor-
responding class, yielding the parameter matrix ˆ =
ˆθi0, ˆθi1
=
( ˆμi0, ˆσi0), ( ˆμi1, ˆσi1)
∀i ∈1, . . . , N.
The prior probability distribution P() includes penali-
sation of differences between the network structure and
a user-defined prior belief B = V × V →[−1, 1], where
the absolute value correlates with the confidence in an
edge. Here we chose B = V × V →{0, 1, −1}, assum-
ing in advance specific activating, inhibiting or missing
edges with maximum confidence. We made use of the
Laplace prior model (laplaceinhib), accounting for both
edge types, i.e. activation and inhibition. The prior belief
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 5 of 21
http://www.biomedcentral.com/1752-0509/8/75
for an edge is defined as P(φij|bij, λ, γ ) =
1
2λe
−
ij
λ , includ-
ing a weighted difference term
ij = |φij −bij|γ with a
weight exponent γ ∈R+. As the edge probabilities are
assumed to be independent, the prior belief for a net-
work structure is derived as the product of those, i.e.
P(|B, λ, γ ) =
i,j
P(φij|bij, λ, γ ), i, j ∈{1, . . . , N}. The
individual edge probabilities lie between 0 and 1
2λ ∀λ, γ ∈
R+. The protein interactions corresponding to our cho-
sen prior are displayed in Additional file 2. The prior’s
impact strength was emphasised in such a way, that only
strongly deviating data influence the network structure,
because the ErbB wiring as well as the MAPK and PI3K
pathways are well examined in literature. This prioritisa-
tion is reflected in the hyperparameter λ set to 0.0001. For
the parameter γ we chose one, neglecting extra penali-
sation of deviation from the prior. These settings should
preserve robustness, but at the same time allow enough
impact strength of strongly differing data values.
The network inference via inhibMCMC spanned 50,000
iterations with the first 25,000 iterative steps as burn-in
phase. To ensure convergence, ten parallel MCMC chains
were run, each initialised with a starting network. Con-
vergence was validated via Gelman diagnostic [49]. Nine
of the initial ten networks were randomly generated, i.e.
for the defined nodes activating, inhibiting or no edges
were sampled. The remaining network assumed no con-
nections between the nodes. These initial networks were
pruned to the following constraints. Firstly, the nodes
related to the growth factors and drugs must not have
any ingoing edges. Above that, the indegree of all nodes
was limited to four. Finally, no self-loops were allowed.
To find significantly occurring edges among the indepen-
dent runs, merging into a consensus network, a Wilcoxon
rank sum testing procedure was used. In detail, in each
run the amount of sampled activations and inhibitions per
edge was counted and divided by the total number of sam-
pled edges. Subsequently the null-hypothesis was tested,
whether the means of these ten edge-specific confidence
values equal the same for activation and inhibition. In case
of not rejecting the null-hypothesis, coming along with an
adjusted p-value exceeding the significance level α = 0.05,
no edge was assumed. Otherwise, the respective alter-
native determined the type of interaction. Adjustment
for multiple testing followed the method of Benjamini
and Hochberg, controlling the false discovery rate [50].
The whole procedure was embedded into a leave-one-
out cross-validation approach. So each of the ten MCMC
chains was left out once, and the testing algorithm was
applied to the remaining runs. An edge was included in
the final consensus network if it occurred in all of the
cross validation runs. Finally, to prevent excessive spuri-
ous or obsolete connections ascribable to transitivity, as
argued by Bo Na Ki et al. [51], newly reconstructed edges
were successively added to the prior network according to
ddepn significance and fit of resulting attractor states to
the observations of Henjes et al. [28].
Perturbation simulations
To figure out which input of drug combination leads to
a certain attractor state of the reconstructed network
system, the R package BoolNet [21] was applied. The moti-
vation was based on the assumption that attractors, rep-
resenting cycles of states, comprise the stable states of cell
function. In those states networks mostly reside. Hence,
they mirror system phenotypes, dependent on the pertur-
bation context. To the best of our knowledge, apart from
BoolNet, there are hardly any R packages offering attractor
calculations for Boolean networks. This package supports
import of networks in form of files containing Boolean
formulas. So it could be easily integrated in our workflow
as subsequent analysis step after network reconstruction.
We used its functionality to identify attractors in a syn-
chronous and an asynchronous way. The resulting attrac-
tors were steady-state attractors. These consist of only
one state, in which all transitions from this state result.
These attractors are identical for synchronous and asyn-
chronous updates. We focused on the steady-states, as
these should reflect the homoeostatic system state of the
cell lines. Intermediate transition states would be interest-
ing as well, but due to the large amount of the involved
targets, it would have been too complex to analyse those
here in detail.
The search started from predefined initial states of
the network nodes. The drug and growth factor nodes
were fixed to specific values, reflecting the conducted
experiment to be simulated. For short-term signalling,
perturbations included all possible combinations of the
therapeutics under the combined stimulus of EGF and
HRG. Although the data of separate stimulation with EGF
and HRG was used for network reconstructions, here we
focused on the combined treatment, representing a more
natural tumour environment than a single growth factor
alone. Two possible binary states, i.e. active (1) or pas-
sive (0), to the power of three different drugs led to eight
possible combinations. These were used as fixed input
conditions, as the effect was assumed to be continuously
valid. Analogously, the growth factors were permanently
fixed to one. The remaining protein activity start states
were initialised with zero. These components were flexi-
ble towards updates. In the long-term measurements, no
growth factors were involved but full growth medium.
This was defined as one stimulating input S, initially acti-
vating the ErbB receptors. This also led to eight fixed input
combinations.
BoolNet expects network representation in form of log-
ical interaction rules as input. In contrast, ddepn deliv-
ers network reconstruction output in terms of adjacency
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 6 of 21
http://www.biomedcentral.com/1752-0509/8/75
matrices. Therefore, we incorporated an interface func-
tion into the ddepn package, called adjacencyMa-
trix_to_logicalRules. In detail, the loadNetwork function
of BoolNet requires a file containing row-wise logical acti-
vation rules of each network node. Each row looks like
‘target node, (activator_1 | activator_2) & !(inhibitor_1 |
inhibitor_2)’, here exemplary for a node with two ingo-
ing activating and inhibiting edges each. The logical OR
operator is encoded by ‘|’, the logical AND is encoded
by ‘&’, and logical negation is represented by ‘!’. Accord-
ingly, all of the A inferred activating nodes V+
=
{va : a ∈1, . . . , A, A < N} of a target node vj, represented
by an adjacency matrix entry φaj = 1, and vj itself were
connected via OR operators. This ensured that at least one
of the activators or the target protein itself had to be active
to activate the target node. Analogously, the I inhibiting
nodes V−= {vi : i ∈1, . . . , I, I < N} with φij = −1 were
connected via OR operators. A logical negation opera-
tor was attached to ensure that the activity of one of the
nodes vi would result in an inactive node vj. Both sets
of activators and negated inhibitors were then connected
via a logical AND operator. After conversion of the adja-
cency matrices to logical rules, those were implemented
in BoolNet into a computational model, to perform per-
turbation simulations per cell line and time course as well
as subsequent analyses of the resulting attractor states.
Results and discussion
The complete workflow, holding for both, short- and
long-term analysis, is depicted in Figure 1. For a better
understanding of the discussion on MAPK and PI3K sig-
nalling, Figure 2 displays the interactions between the
main MAPK and PI3K targets of the ErbB prior net-
works. It shows the preferred pathway activations by all
possible homo- and heterodimers formed upon ligand
binding to the ErbB-1 and ErbB-3 receptors [9,29,52-54].
The confidence values, representing the likeliness of the
reconstructed network edges, are shown in Additional
file 4.
Short-term signalling network reconstruction
The short-term signalling networks, reconstructed by the
ddepn algorithm, are depicted in Figure 3. The equivalent
Boolean logical interaction rules are listed in Additional
file 5. In comparison to the prior network, newly inferred
edges were specific for each cell line, and all of them
were activating. For HCC1954 and BT474, seven addi-
tional edges were reconstructed, while in SKBR3 only two
new edges were reconstructed. No prior edge deletion or
type reversal took place.
HCC1954 is driven by the PI3K as well as the MAPK pathway
In HCC1954, the new edges contributed to both, PI3K
and MAPK, signalling. The interaction ErbB-1→ErbB-2
reflected a dominant role of heterodimerisation of both
receptors, as described by Henjes et al. [28]. The fact
that it was specifically inferred for HCC1954, pointed
to hyperactive ErbB-1/2 heterodimers here. These are
known to trigger the MAPK but also, to a lesser extent,
the PI3K pathway. The link PDK1→MEK1/2, supported
by Sato et al. [55], stressed crosstalk between these path-
ways, placing PDK1 into a key position in the PI3K
pathway, and MEK1/2 in the MAPK pathway, respec-
tively. Two of the new edges in HCC1954, PDK1→ErbB-2
and p70S6K→AKT, contributed to feedback loops, which
were not present in the other two cell lines. Such a
topological network element could stabilise the known
trastuzumab resistance by boosting the oncogenic effect
of ErbB-2 and the mutant hyperactive PI3K pathway. Evi-
dence for the feedback mechanism involving PDK1 was
provided by Maurer et al. [56] and Tseng et al. [57]. Vega
et al. noted an indirect activation of AKT by p70S6K via
mTOR [58].
BT474 is driven by the PI3K pathway, while SKBR3 is driven by
the MAPK pathway
Comparably to HCC1954, in BT474 an edge indicat-
ing hyperactive heterodimers was found, namely ErbB-
3→ErbB-2, here interestingly with a strong impact on
AKT [28]. BT474 is known to contain a rare type
of PIK3CA mutation [43]. Pathway crosstalk was also
observed in BT474, but here MEK1/2 activated PDK1, and
not vice versa like in HCC1954. This edge was supported
by Frödin et al. [59], underlining dominant PI3K signalling
in this cell line.
The newly detected interactions in SKBR3 started from
ErbB-3 and PDK1, and both activated ERK1/2. This
reflected a dominant MAPK pathway, in which ErbB-
3→ERK1/2 was interpretable as indirect stimulation of
ERK1/2 via MEK1/2, activated by ErbB-2/3 dimers [55].
Perturbation simulations on short-term networks
Perturbations included all possible combinations of
the therapeutics erlotinib, pertuzumab and trastuzumab
under combined stimulation of EGF and HRG. All
inferred attractors were simple and consisted of one
steady-state. This means that all transitions from this state
result in the state itself. Table 1 summarises all simula-
tion outcomes for the attractors of the AKT and ERK1/2
proteins, as those are key players in the PI3K (AKT) and
MAPK (ERK1/2) pathways. Additionally, the results for
p70S6K are listed there, as both pathways regulate this
protein [60].
Stimulation with EGF and HRG should result in acti-
vation of ErbB-1 and ErbB-3, followed by dimerisation
amongst ErbB members. This should initialise signalling
cascades in the MAPK and PI3K pathways (Figure 2).
Indeed, AKT, ERK1/2 and p70S6K got activated in
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 7 of 21
http://www.biomedcentral.com/1752-0509/8/75
Figure 1 Modelling workflow. The figure summarises the applied modelling approach. RPPA data of three individual breast cancer cell lines were
generated under short- and long-term drug treatment. They constituted the basis for network reconstruction in combination with prior literature
knowledge about protein wiring. The reconstructed networks per cell line and time course in turn underwent Boolean perturbation simulations to
reveal optimal drug treatments.
all cell lines, which was revealed by simulations as
well as observations in graphical analyses (Table 1,
Figures 4, 5, 6).
As we were interested in identifying optimal drug treat-
ments, Table 2 summarises the corresponding statistical
results. Most of them were supported by the perturbation
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 8 of 21
http://www.biomedcentral.com/1752-0509/8/75
Figure 2 Scheme of ErbB dimers related MAPK and PI3K pathway activation. The figure depicts the different homo- and heterodimers of ErbB
receptors, induced upon activation via the ligands EGF or HRG. The active dimers then initialise the MAPK and PI3K signalling cascades. The orange
(PI3K) and green (MAPK) arrows denote, which dimer activates which pathway.
simulation results, corresponding to attractor states of
AKT, ERK1/2 and p70S6K being zero. Four main con-
clusions were drawn from these results, which will be
discussed in detail in the following subsections. Firstly,
inhibition of PI3K signalling, reflected by downregulated
AKT, required the combined treatment with erlotinib,
pertuzumab and trastuzumab. Secondly, inhibition of the
MAPK pathway, represented by ERK1/2, was reached
with erlotinib alone in SKBR3 and HCC1954. BT474 addi-
tionally needed pertuzumab. Thirdly, the protein activity
of p70S6K was influenced by both, PI3K and MAPK,
pathways. The drug response differed between cell lines,
indicating both pathways contribute to a different extent.
Finally, the drug effect on PI3K signalling was much better
in SKBR3 than in HCC1954, pointing to resistance in the
latter cell line.
Inhibition of PI3K signalling requires drug combinations
In SKBR3, the triple drug combination was most effec-
tive in inhibiting AKT (Figure 4, Table 2). In BT474,
pertuzumab combined with erlotinib was most effi-
cient, but AKT signalling was not fully suppressed as
in SKBR3 (Figure 5). Statistically, we did not infer any
significant positive drug effect in this cell line. Obvi-
ously, erlotinib in synergistic combination with at least
pertuzumab was needed to block the ErbB-2 receptor
and its heterodimerisation, mainly with ErbB-1, but also
ErbB-3. The HRG activated ErbB-2/3 heterodimers and
PI3K pathway in BT474, as revealed by the network
reconstructions, might have prevented a potent drug
efficacy.
Interestingly, BT474 and SKBR3 required pertuzumab.
This drug was especially designed to prevent het-
erodimerisation with ErbB-2. The stimuli EGF and HRG
together activate PI3K signalling by ErbB-2/3, ErbB-
1/2 and ErbB-1/3 dimers (Figure 2). The need for
pertuzumab combined with erlotinib indicated an impor-
tant role of ErbB-1/2 dimers. This was supported by the
fact, that in HCC1954 with dominant heterodimers of
this type, as revealed by network reconstructions, none
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 9 of 21
http://www.biomedcentral.com/1752-0509/8/75
Figure 3 Reconstructed short-term signalling networks. The figure displays the reconstructed short-term signalling networks coloured
according to the preserved prior reference network (black) and newly inferred (added) individual edges per cell line. Target proteins are represented
as rectangles with stimuli and drugs coloured in red. The three drug names erlotinib, trastuzumab and pertuzumab are abbreviated via their first
letters. Solid arrows denote activating interactions while dashed ones represent inhibitions.
Table 1 Attractor states of short-term perturbation simulations
BT474
HCC1954
SKBR3
Simulation
AKT
ERK1/2
p70S6K
AKT
ERK1/2
p70S6K
AKT
ERK1/2
p70S6K
A
E
A
E
A
E
A
E
A
E
A
E
A
E
A
E
A
E
X
1
1
1
1
1
1
1
1
1
E
0
1
1
0
1
0
1
0
1
0
1
0
0
1
P
1
1
1
0
1
0
1
0
1
0
1
0
1
0
1
T
1
1
1
1
1
1
1
1
1
E, P
1
1
0
1
0
1
0
0
1
0
0
0
0
1
E, T
0
1
1
0
1
1
0
1
1
0
1
0
0
1
P, T
1
1
1
0
1
0
0
1
0
0
0
1
E, P, T
1
1
0
1
0
1
0
0
1
0
0
0
The therapeutics erlotinib, trastuzumab and pertuzumab, abbreviated by first letters, that were permanently active besides EGF and HRG in the simulated
perturbation conditions are stored in the column Simulation. No simulated drug treatment is denoted by ‘X’. The A columns hold the attractor states of the proteins
AKT, ERK1/2 and p70S6K, associated with the perturbations. The E columns contain the protein activity status, statistically deduced from the experimental data. In case
of a significant (p-value < 0.05) combined influence of both, drug treatment and time, on the protein signal intensity, a Wilcoxon rank sum test was conducted for the
measurements at time point 60 minutes. The drug treatments leading to significantly (p-value < 0.05) smaller intensity values compared to the control measurement
‘X’ were considered as efficient inhibitors, resulting in a table entry of zero. Consistency between simulations and experimental observations is printed in bold.
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 10 of 21
http://www.biomedcentral.com/1752-0509/8/75
Figure 4 SKBR3 short-term time courses of AKT, ERK1/2 and p70S6K. The figure shows splines and related standard error bars of the measured
RPPA data for AKT, ERK1/2 and p70S6K after combined EGF and HRG stimulation in the SKBR3 cell line. The measurements included ten time points
up to 60 minutes. The different drug treatments are marked by different colours with ‘X’ denoting no drug treatment.
of the drugs was likewise efficient in inhibiting AKT
(Figure 6). However, the optimal effect was revealed for
the triple drug combination (Table 2). The simulations
suggested pertuzumab alone or a combination of both
monoclonal antibodies (Table 1). It has to be kept in
mind, that the attractor states resembled a long-term
steady state, which can differ from observations up to 60
minutes.
The perturbation simulations in BT474 did not lead to
inactive AKT upon combined pertuzumab and erlotinib
treatment. Instead, erlotinib alone or combined with
trastuzumab was efficient (Table 1). Nevertheless, this
supported the need for the small molecule inhibitor
and a monoclonal antibody to suppress ErbB-2 induced
PI3K signalling. In SKBR3, the attractor states con-
firmed the described optimal drug treatment to deactivate
AKT. Trastuzumab, when applied alone, was the only
treatment without a positive effect in the simulations
(Table 1).
Inhibition of MAPK signalling requires erlotinib
Signalling through the MAPK pathway, represented by
ERK1/2 activation, was efficiently inhibited by erlotinib
alone in both, HCC1954 (Figure 6, Table 2) and SKBR3
(Figure 4, Table 2), cell lines. EGF activates the MAPK
pathway via ErbB-1 homodimers and ErbB-1/2 het-
erodimers (Figure 2). Both are prevented by ErbB-1 inhibi-
tion via erlotinib, which was especially designed to target
this receptor.
In BT474, pertuzumab plus erlotinib was required
(Figure 5, Table 2). This was analogous to the situation in
PI3K signalling.
HRG activates the MAPK pathway via ErbB-2/3 het-
erodimers (Figure 2). Obviously, BT474 needed the
addition of the monoclonal antibody due to dominant
ErbB-2/3 formation and activity. On the contrary, the
other two cell lines just needed erlotinib alone. Here, in
addition to the ErbB-1 dimers, the ligand-independent
ErbB-2 homodimers might have driven ERK1/2 activation
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 11 of 21
http://www.biomedcentral.com/1752-0509/8/75
Figure 5 BT474 short-term time courses of AKT, ERK1/2 and p70S6K. The figure shows splines and related standard error bars of the measured
RPPA data for AKT, ERK1/2 and p70S6K after combined EGF and HRG stimulation in the BT474 cell line. The measurements included ten time points
up to 60 minutes. The different drug treatments are marked by different colours with ‘X’ denoting no drug treatment.
and could be inhibited by the small molecule inhibitor.
Efficacy of erlotinib towards ErbB-2 dimers was previ-
ously mentioned by Schaefer et al. [61].
In BT474, the simulations resulted in active ERK1/2
states, resisting drug treatment (Table 1). In HCC1954
and SKBR3, the positive effect of erlotinib was supported
by the simulations. The attractor states were additionally
inactive for all other (combinatorial) drug treatments, but
not trastuzumab alone.
p70S6K is influenced by both, PI3K and MAPK, pathways
The target p70S6K is upstream influenced by the PI3K
as well as the MAPK pathway (Figure 2). Hence, p70S6K
merges both pathways, leading to activation of RPS6 [60].
The three cell lines showed different pathway prefer-
ences. BT474 required the combination of pertuzumab
and erlotinib to suppress p70S6K (Figure 5). On the con-
trary, in SKBR3 the triple drug combination was shown to
be optimal (Table 2). Obviously, the effect was driven by
erlotinib (Figure 4), which was supported by the attractor
states of p70S6K (Table 1). This resembled the drug
response of ERK1/2 and reflected a stronger influence by
the MAPK pathway. In HCC1954, deactivation of p70S6K
was reached via application of erlotinib combined with
pertuzumab (Table 2). The treatment with erlotinib alone
had a similar effect (Figure 6), while the simulations just
confirmed a positive effect of pertuzumab (Table 1). Thus,
this cell line seemed to be influenced by both, PI3K and
MAPK, pathways.
These results were in line with the newly inferred edges
in the network reconstructions. They pointed to a strong
influence of PI3K in BT474 in contrast to a dominant
MAPK pathway in SKBR3. HCC1954 was influenced by
both pathways to a similar extent.
To follow up on the hypothesis that different path-
ways contribute to a different extent in individual cell
lines, we tested correlation between the p70S6K time
course and the ones of AKT and ERK1/2, respectively.
In BT474, p70S6K correlated positively with AKT (p-
value 0.01, Kendall’s τ estimate 0.64). In HCC1954,
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 12 of 21
http://www.biomedcentral.com/1752-0509/8/75
Figure 6 HCC1954 short-term time courses of AKT, ERK1/2 and p70S6K. The figure shows splines and related standard error bars of the
measured RPPA data for AKT, ERK1/2 and p70S6K after combined EGF and HRG stimulation in the HCC1954 cell line. The measurements included
ten time points up to 60 minutes. The different drug treatments are marked by different colours with ‘X’ denoting no drug treatment.
p70S6K correlated positively with both, AKT (p-value <
2.22 · 10−16, Kendall’s τ estimate 0.69) and ERK1/2
(p-value < 2.22 · 10−16, Kendall’s τ estimate 0.87). In
SKBR3, p70S6K also correlated positively with both, AKT
(p-value 0.05, Kendall’s τ estimate 0.51) and ERK1/2
(p-value 0.02, Kendall’s τ estimate 0.6), with a stronger
tendency towards MAPK signalling. The correlation
was not as convincing as in the other two cell lines.
One could speculate, that the dominance of the MAPK
pathway in SKBR3 cells was not as strong as the domi-
nance of the PI3K pathway in BT474. This was supported
by the reconstructed networks. They revealed down-
Table 2 Optimal drug treatment in short-term signalling
Cell line
AKT
ERK1/2
p70S6K
BT474
-
PE
PE
HCC1954
PTE
E
PE
SKBR3
PTE
E
PTE
The table summarises the optimal drug treatments for the short-term data, leading to inactive AKT, ERK1/2 and p70S6K, respectively. In case of a significant (p-value <
0.05) combined influence of both, drug treatment and time, on the protein signal intensity, a Wilcoxon rank sum test was conducted for the measurements at time
point 60 minutes, testing for significantly (p-value < 0.05) smaller intensity values under the drug treatment compared to the control measurement. The drug
treatment with the smallest p-value was considered as the optimal inhibitor. No inferred significant positive drug effect is denoted by ‘-’. The therapeutics erlotinib,
trastuzumab and pertuzumab are abbreviated by their first letters. More than one letter denotes drug combinations. The growth factors EGF and HRG were added in
combination to the cell lines and permanently active in the simulated perturbation conditions. The column Cell line holds the cell lines under consideration. The
columns AKT, ERK1/2 and p70S6K hold the optimal drug combinations for each target. If those were confirmed by the attractor states (0) of perturbation simulations,
they are printed in bold.
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 13 of 21
http://www.biomedcentral.com/1752-0509/8/75
stream effects of MAPK signalling in SKBR3, while they
revealed hyperactive ErbB-2/3 dimers in BT474. The
dimers drive PI3K already at the receptor layer, and espe-
cially ErbB-2/3 dimers are regarded as the most potent
heterodimer [29].
Drug resistance in HCC1954 regarding the PI3K pathway
In HCC1954, the inferred optimal treatment against AKT
signalling with the triple drug combination was not con-
vincing (Figure 6). Analogously, Henjes et al. did not
monitor any positive drug effect on AKT under EGF appli-
cation alone [28]. However, the simulations suggested
pertuzumab alone or a combination of both monoclonal
antibodies to inhibit AKT phosphorylation. In principle,
divergence of simulations from experimental observations
can be expected, as the simulated steady state of the sys-
tem does not necessarily have to be reached after the
measured period of time. Anyhow, the apparent resis-
tance here pointed to a hyperactive PI3K pathway which
was explainable by the newly inferred HCC1954 edges
described in the previous subsection. They represented
feedback loops, hyperactive ErbB-1/2 heterodimers and
pathway crosstalk. On the contrary, in SKBR3, the triple
drug combination worked well, as described before. The
simulations even predicted efficacy of every other drug
(combination) apart from trastuzumab alone. The drug
efficacy towards AKT in this cell line could be explained
by the fact that the two reconstructed interactions in
SKBR3 mainly promoted the MAPK instead of the PI3K
pathway.
The regulation of AKT activity under drug influence,
highly diverging in HCC1954 and SKBR3, attracted our
attention. Therefore we intended testing for edgetic muta-
tions, as discussed by Zhong et al. [62], leading to AKT
gain-of-function in HCC1954. Such mutations, perturb-
ing not a node but an edge of a network, are speculated to
have deeper impact on phenotypic manifestation of a dis-
ease. In detail, we removed each of the AKT stimulating
edges outgoing from p70S6K, PDK1, mTOR and ErbB-
3, alone or in all possible eleven combinations. We then
computed the attractor states for the modified networks
in HCC1954.
Removal of the connections of mTOR, PDK1 and ErbB-3
alone or combined had no influence on improving drug
effects, i.e. AKT just got inactive under pertuzumab treat-
ment. Involvement of p70S6K→AKT in the withdrawal
process led to much better results. Removed alone or in
double combinations with the aforementioned edges, as
well as in the two triple combinations containing mTOR,
AKT was deactivated under all drug treatments, but not
yet trastuzumab alone. Finally, simultaneous removal of
the outgoing connections from p70S6K, ErbB-3 and PDK1
with or without mTOR, turned out as the only combina-
tion enabling potency of all possible drug combinations,
including trastuzumab alone. This hinted at a less strong
impact of mTOR on AKT here, but indicated synergistic
drug resistance potential of p70S6K, ErbB-3 and PDK1,
also due to the newly inferred edges.
Long-term signalling network reconstruction
The reconstructed long-term signalling networks per cell
line are displayed in the Additional file 6. Additional
file 5 lists the equivalent Boolean logical interaction
rules. Compared to the prior network, most of the
newly inferred edges were individual for each cell line,
but HCC1954 shared ErbB-1→ERK1/2 with SKBR3, for
example. This seemed to be an indirect edge via cRAF,
as represented in the prior network. Besides activat-
ing connections, also inhibiting ones and edge deletions
occurred. For HCC1954, ten new interactions were recon-
structed, while two were deleted. In BT474, nine new
links were added, and one edge was deleted. In SKBR3,
we inferred 20 new connections and one deletion, namely
the removal of p53 activation via p38, bearing oncogenic
risk [63,64].
In contrast to the short-term networks, new feedback
loops were reconstructed in every cell line, not exclu-
sively in HCC1954. In HCC1954, the mutual activation
between p53 and RB established such a feedback mecha-
nism. For SKBR3 we even inferred two edges, each form-
ing feedback loops. Contrary to HCC1954, p53 inhibited
RB. The second loop connection was inhibition of ErbB-
3 by AKT, pointing to a negative feedback against PI3K
signalling [65-67].
In HCC1954, the newly inferred edges Cyclin B1→AKT
and ErbB-3→ErbB-1 contributed to PI3K signalling, of
which the latter was explainable as heterodimers. The
newly inferred edge cJUN→ErbB-1 in HCC1954 also
indicated raised activity of ErbB-1. Interestingly, in SKBR3
we conducted an inhibiting edge from Cyclin B1 to AKT
but instead an activating one to ERK1/2, contributing to
MAPK signalling, which was also stated by Abrieu et al.
[68]. Another new edge in HCC1954 involved a cell cycle
player, i.e. activation of Cyclin D1 by p70S6K [69]. Accord-
ingly, we inferred RPS6→Cyclin D1 in BT474, with RPS6
as downstream target of p70S6K. In SKBR3, the edge
p70S6K→Cyclin B1 was reconstructed. A further inter-
esting new activating edge in HCC1954 led from RB to
TSC2, while we inferred a reversed inhibition in SKBR3.
Searle et al. discussed targeting RB deficient cancers by
deactivating TSC2 [70].
Two novel interactions in BT474 activated Cyclin B1,
arising from ErbB-1 and ErbB-3, respectively, which
meant that mitosis was driven by ErbB-1/3 dimers in this
cell line. This indicated a hyperactive PI3K pathway, as
revealed in the short-term case.
In SKBR3, we reconstructed an outgoing edge from
the artificial network stimulus S, representing full growth
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 14 of 21
http://www.biomedcentral.com/1752-0509/8/75
medium, activating AKT. This could be explained as
strong activation of AKT, driving PI3K signalling in
this cell line. The new edges ErbB-2→TSC2 and ErbB-
3→PRAS had to be interpreted as indirect effects, too.
They pointed to activity of ErbB-2/3 dimers, feeding
into both, MAPK and PI3K, pathways. The edge ErbB-
2→TSC2 could imply an oncogenic role of TSC2. Liu et al.
discussed a context dependent functionality of TSC2 [71].
Perturbation simulations on long-term networks
Similarly to the perturbation simulations for the short-
term networks, we performed those for the long-term
networks under all eight initial state combinations of the
therapeutics erlotinib, pertuzumab and trastuzumab. Also
here, all inferred attractors were simple and consisted of
one steady-state. Table 3 contains the simulation results
for the attractors of the RPS6 and RB proteins, as those
are key players in cell growth and proliferation and mainly
comparable to the experimental results of Henjes et al.
[28] for HCC1954 and SKBR3. We analysed the attractor
states of AKT and ERK1/2, too, but the results are not
explicitly listed, since they mostly resembled the ones of
RPS6.
The control measurements without any drug treatment
should result in activation of ErbB members and dimeri-
sation events, promoting cell growth and proliferation. In
fact, this was expressed as reasonable activation of AKT,
ERK1/2 and RPS6 in all cell lines, which held for simu-
lations as well as experimental observations. In contrast,
the attractor states of RB were inactive in all cell lines
(Table 3). Actually, a continuously rising stimulation effect
over 30 hours was not observed for HCC1954 and SKBR3
by Henjes et al. [28] either.
The attractor states of RPS6 and RB were identical in all
cell lines (Table 3). All drugs, except trastuzumab under
stimulation alone, led to inactive attractor states of RPS6.
This was also the case for ERK1/2 in all cell lines, as well
as AKT in BT474 and HCC1954. In SKBR3, the attractor
states of AKT were just inactive without the stimulus. All
therapeutics, including trastuzumab, resulted in deacti-
vated attractor states of RB. The statistically inferred drug
effects for AKT, ERK1/2, RB and RPS6 were slightly differ-
ent. Table 4 summarises the optimal drug combinations,
confirming and extending the observations of Henjes
et al. [28]. Most of them were supported by the perturba-
tion simulation results, corresponding to attractor states
of AKT, ERK1/2, RB and RPS6 being zero.
The optimal long-term drug response for AKT and ERK1/2
confirms short-term observations
As shown in Figure 7, the best drug response in BT474
and HCC1954 regarding AKT was yielded for a combina-
tion of trastuzumab and erlotinib. Statistically, we inferred
no positive effect in BT474 at all, which is explainable
by the fact that we just considered a combined effect of
drug treatment and time. Although the time courses of
AKT signalling with and without the drug treatment were
differing in the intensity strength, the signalling profiles
were similar. This parallel shift indicated no time effect.
Instead, the group effect was significant (p-value < 2 ·
10−16). This was also the explanation, why we detected
erlotinib, but not the combination with trastuzumab, as
the optimal treatment in HCC1954 (Table 4). In SKBR3,
we inferred the triple drug combination as the optimal
one, but the combination of both monoclonal antibodies
alone also had a significant effect over time (Figure 7).
Table 3 Attractor states of long-term perturbation simulations
BT474
HCC1954
SKBR3
Simulation
RPS6
RB
RPS6
RB
RPS6
RB
A
E
A
E
A
E
A
E
A
E
A
E
X
1
0
1
1
0
1
1
0
1
E
0
1
0
0
1
0
0
1
0
P
0
-
0
-
0
-
0
-
0
-
0
-
T
1
-
0
-
1
-
0
-
1
-
0
-
E, P
0
1
0
0
1
0
0
1
0
E, T
0
1
0
0
1
0
0
0
P, T
0
0
0
1
0
1
0
0
E, P, T
0
1
0
0
1
0
1
0
0
The therapeutics erlotinib, trastuzumab and pertuzumab, abbreviated by first letters, that were permanently active in the simulated perturbation conditions besides
the stimulus S, standing for the full growth medium, are stored in the column Simulation. No simulated drug treatment is denoted by ‘X’. The A columns hold the
attractor states of the proteins RPS6 and RB associated with the perturbations. The E columns contain the protein activity status, statistically deduced from the
experimental data. In case of a significant (p-value < 0.05) combined influence of both, drug treatment and time, on the protein signal intensity, a Wilcoxon rank sum
test was conducted for the measurements at time point 30 hours. The drug treatments leading to significantly (p-value < 0.05) smaller intensity values compared to
the control measurement ‘X’ were considered as efficient inhibitors, resulting in a table entry of zero. Lacking comparable experiments is labelled as ‘-’, while
consistency between simulations and experimental observations is printed in bold.
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 15 of 21
http://www.biomedcentral.com/1752-0509/8/75
Table 4 Optimal drug treatment in long-term signalling
Cell line
AKT
ERK1/2
RB
RPS6
BT474
-
TE
E
TP
HCC1954
E
TE
E
-
SKBR3
PTE
TE
TE
TE
The table summarises the optimal drug treatments for the long-term data,
leading to inactive AKT, ERK1/2, RB and RPS6, respectively. In case of a significant
(p-value < 0.05) combined influence of both, drug treatment and time, on the
protein signal intensity, a Wilcoxon rank sum test was conducted for the
measurements at time point 30 hours, testing for significantly (p-value < 0.05)
smaller intensity values under the drug treatment compared to the control
measurement. The drug treatment with the smallest p-value was considered as
the optimal inhibitor. No inferred significant positive drug effect is denoted by ‘-’.
The therapeutics erlotinib, trastuzumab and pertuzumab are abbreviated by
their first letters. More than one letter denotes drug combinations. The column
Cell line holds the cell lines under consideration. The columns AKT, ERK1/2, RB
and RPS6 hold the optimal drug combinations for each target. If those were
confirmed by the attractor states (0) of perturbation simulations, they are
printed in bold.
Hence, like in the short-term results, a drug combination
was required to suppress PI3K signalling, here with an
obvious need for trastuzumab. For BT474 and HCC1954,
this was supported by the simulation results, in which
trastuzumab alone had no effect, but was efficient within
drug combinations. In HCC1954, even the best drug
response was not convincing (Figure 7), pointing to a
dominant PI3K pathway, as revealed in the short-term
analysis.
Interestingly, SKBR3 showed a strong activation peak of
AKT phosphorylation between 8 and 18 hours (Figure 7),
which was just suppressed under combined application of
trastuzumab and pertuzumab. We revealed a positive cor-
relation with ERK1/2 (p-value 0.02, Kendall’s τ estimate
0.6) and RPS6 (p-value 0.01, Kendall’s τ estimate 0.64).
The reconstructed edges S→AKT and ErbB-1→ERK1/2
in SKBR3 indicated strong activation of AKT and ERK1/2.
In addition to the prior network, in which AKT and
ERK1/2 fed into RPS6 phosphorylation via p70S6K, some
of the novel edges pointed to a positive feedback from
p70S6K or RPS6 to ERK1/2. The feedback from p70S6K
via Cyclin B1, for example, was expressed by the edges
p70S6K→Cyclin B1 and Cyclin B1→ERK1/2. Compared
to the short-term results, indicating a dominant MAPK
pathway, this long-term observation indicated strong sig-
nalling via both, PI3K and MAPK, pathways in SKBR3.
As displayed in Figure 8, erlotinib alone or in combina-
tion with trastuzumab showed the optimal effect against
ERK1/2 in all of the three cell lines. This was in line
with the short-term observations, and confirmed by the
Figure 7 Long-term time courses of AKT for all cell lines. The figure shows splines and related standard error bars of the measured RPPA data for
AKT in all cell lines. The measurements included ten time points up to 30 hours. The different drug treatments are marked by different colours with
‘X’ denoting no drug treatment.
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 16 of 21
http://www.biomedcentral.com/1752-0509/8/75
Figure 8 Long-term time courses of ERK1/2 for all cell lines. The figure shows splines and related standard error bars of the measured RPPA data
for ERK1/2 in all cell lines. The measurements included ten time points up to 30 hours. The different drug treatments are marked by different colours
with ‘X’ denoting no drug treatment.
perturbation simulations. Statistically, the most potent
drug effect was yielded with the combination of erlotinib
and trastuzumab (Table 4).
Quick drug response for RPS6 and delayed response for RB
As shown in Figure 9, in BT474, the simulation based
predicted efficacy of erlotinib alone to counteract RPS6
(Table 3) was not as convincing as in case of drug combi-
nations. A combination of pertuzumab and trastuzumab
worked best (Table 4). For RB, the simulated drug
effects in BT474 resembled the observed ones (Table 3,
Figure 10), with a positive effect of all measured drug
treatments. Erlotinib was inferred as the optimal treat-
ment (Table 4). Though, the drug impact unfolded not
before 18 hours.
In HCC1954, it was the combination of both mon-
oclonal antibodies, that failed in deactivating RPS6
(Figure 9), while the simulations predicted trastuzumab
alone to fail (Table 3). The graphical observations were
similar for RB (Figure 10). The newly inferred edges ErbB-
3→ErbB-1 and cJUN→ErbB-1 in HCC1954 explained the
necessity for erlotinib against ErbB-1 dimers. The positive
impact of erlotinib, the optimal treatment against RB sig-
nalling (Table 4), was supported by simulations. However,
it did not unfold before 12-18 hours, in case of RB as
well as RPS6. Regarding RPS6, no significant effect was
detected for HCC1954 (Table 4).
According to Henjes et al. [28], in SKBR3 erlotinib and
all therapeutic combinations helped to suppress RPS6,
which was supported by the simulations (Table 3). As
shown in Figure 9, the combination of trastuzumab and
erlotinib was the only one, that revealed its continuous
inhibiting effect already after one hour. This combined
treatment was also statistically inferred as the optimal one
(Table 4). The same combination was optimal with respect
to RB activity, which was also in line with the simulations.
Here, analogously to BT474 and HCC1954, the drug effect
did not appear before 18 hours (Figure 10).
As the combination of trastuzumab and erlotinib was
efficient in all of the three cell lines against RPS6 as well
as RB phosphorylation, we further analysed target correla-
tions under this drug combination to explain the different
rapidness of drug responses.
In BT474, RB positively correlated with Cyclin B1 (p-
value 0.02, Kendall’s τ estimate 0.6), while RPS6 positively
correlated with ERK1/2 (p-value 0.02, Kendall’s τ esti-
mate 0.6). Obviously, RPS6 was mainly stimulated by
the MAPK pathway, which was efficiently inhibited by
the combination of trastuzumab and erlotinib in a fast
manner. On the contrary, RB seemed to be influenced
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 17 of 21
http://www.biomedcentral.com/1752-0509/8/75
Figure 9 Long-term time courses of RPS6 for all cell lines. The figure shows splines and related standard error bars of the measured RPPA data
for RPS6 in all cell lines. The measurements included ten time points up to 30 hours. The different drug treatments are marked by different colours
with ‘X’ denoting no drug treatment.
by Cyclin B1. The newly reconstructed edges ErbB-
1→Cyclin B1 and ErbB-3→Cyclin B1 supported hyper-
activity of Cyclin B1, driven by ErbB-1/3 heterodimers.
In SKBR3, RB negatively correlated with PRAS (p-value
0.05, Kendall’s τ estimate -0.51) and TSC2 (p-value 0.03,
Kendall’s τ estimate -0.56), while RPS6 positively corre-
lated with AKT (p-value 0.03, Kendall’s τ estimate 0.56)
and ERK1/2 (p-value 0.02, Kendall’s τ estimate 0.6). Obvi-
ously, like in BT474, RPS6 was mainly activated through
the MAPK pathway. Interestingly, RB seemed to require
inhibition via PRAS or TSC2. The latter was confirmed
via one of the novel edges in SKBR3, namely inhibition of
RB by TSC2. In addition, PRAS as well as TSC2 seemed to
be especially active in this cell line with regard to the new
edges ErbB-3→PRAS and ErbB-2→TSC2.
In HCC1954, the drug response was not only delayed
for RB, but also for RPS6, which was in line with the posi-
tive correlation with RB (p-value < 2.22 · 10−16, Kendall’s
τ estimate 0.73). Like in BT474, Cyclin B1 seemed to be a
driving force, since both, RPS6 (p-value 0.03, Kendall’s τ
estimate 0.56) and RB (p-value < 2.22 · 10−16, Kendall’s
τ estimate 0.82) positively correlated with this target. The
new edge Cyclin B1→AKT supported special activation
of RPS6 via PI3K signalling, leading to a delayed drug
response. Interestingly, we revealed negative correlations,
as observed for SKBR3. In HCC1954, RPS6 and RB corre-
lated with BAX (p-value 0.03, Kendall’s τ estimate -0.56)
and FoxO1/3a (p-value 0.05, Kendall’s τ estimate -0.51),
pointing to a delayed inhibition of RPS6 and RB via BAX
or FoxO1/3a.
Conclusions
Using a combination of reverse and forward engineer-
ing techniques, we focused on deregulated protein inter-
actions in the ErbB network in a Boolean modelling
framework. The reconstructed hypothetical networks
revealed individual protein interactions contributing to
signalling pathway preferences as well as drug resistance
via feedback loops, pathway crosstalk or hyperactive het-
erodimers. While this reverse engineering focused on
the network edges, we concentrated in the subsequent
forward engineering step on the network nodes. The per-
turbation simulations for AKT, ERK1/2, RB and RPS6
mainly confirmed our graphical and statistical analy-
ses as well as the observations of Henjes et al. [28]
regarding (combinatorial) drug efficacy. However they
have to be interpreted as an independent, more prospec-
tive investigation, because stable system states do not
necessarily have to be reached in temporally limited
observations.
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 18 of 21
http://www.biomedcentral.com/1752-0509/8/75
Figure 10 Long-term time courses of RB for all cell lines. The figure shows splines and related standard error bars of the measured RPPA data for
RB in all cell lines. The measurements included ten time points up to 30 hours. The different drug treatments are marked by different colours with ‘X’
denoting no drug treatment.
In the first step, the combined Boolean modelling
approach revealed the mechanisms underlying individual
drug response. In the second step, it predicted the net-
work propagation effects on protein activity, and hence
the drug response itself.
One major finding is, that different breast cancer pheno-
types seem to be driven by specific pathway preferences in
the ErbB network. This leads to individual drug response,
requiring different therapeutic treatments. The perturba-
tion simulations revealed a more diverse drug response
in short-term than in long-term signalling, which stresses
the importance of early intervention at the top level layer
of the signalling network.
Another interesting aspect is to combine edge and
node perturbations in Boolean network models to reveal
edgetic mutations, as we did in the HCC1954 cell line for
AKT.
Basic molecular research, embedded in a Boolean mod-
elling framework here, composes a first step to gain insight
into individual mechanisms of drug response or resistance
mechanisms in breast cancer. Especially, the proteomic
signalling interplay directly effects tumour development
and represents a promising target in cancer therapy, which
has to be understood in more detail in the future.
Additional files
Additional file 1: Proteins and phosphorylation sites involved in
RPPA measurements. The tables show the proteins and phosphorylation
sites involved in RPPA short- and long-term measurements. The antibody
catalogue numbers and providing companies are mentioned in brackets.
For BT474, no experimental short-term data under EGF or HRG stimulation
were available for PDK1. In case of total protein measurements, the column
Phosphosite remains empty (‘-’) apart from the antibody number and
supplier name.
Additional file 2: Literature references for the prior networks of
short- and long-term signalling. The interactions between proteins are
listed line by line in the tables. The column Protein denotes the source of
the connection with the sink called Target. The interaction (Type) is
encoded numerically, i.e. activation is marked by 1, while inhibition is
labelled with 2, e.g. AKT activates mTOR. The column Reference specifies
the supportive publication.
Additional file 3: Workflow of MCMC-based network structure
inference. The inhibMCMC procedure of the ddepn package was run with
maxIter = 50, 000 in 10 parallel runs. The results of the 25,000 iterations
after the burn-in phase were merged into one consensus network. It was
applied for short- and long-term data separately per cell line, leading to six
consensus networks. The figure is based on [6,47].
Additional file 4: Edge confidences for the reconstructed networks of
the three cell lines per time course. In each of the ten MCMC runs,
activation and inhibition edges were sampled. The percentage, i.e. the
confidence, of sampled activation (red) and inhibition (blue) edges in the
25,000 iterations after the burn-in phase are depicted in the boxes. The sink
nodes are displayed in each panel, while the activating, inhibiting or
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 19 of 21
http://www.biomedcentral.com/1752-0509/8/75
missing influence of the source nodes is shown column-wise in the red,
blue or missing boxes. The source node names are displayed at the x-axis
with additional indicators, where ‘-’ refers to an inhibiting influence and ‘+’
is related to activation. The x-axis of the short-term plots is labelled as ‘AKT,
E, EGF, ERBB1, ERBB2, ERBB3, ERK1/2, HRG, MEK1/2, mTOR, P, p70S6K, PDK1,
PKCα, PLCγ , T’. The x-axis of the long-term plots is labelled as ‘AKT, BAX,
cJUN, cRAF, CyclinB1, CyclinD1, E, ERBB1, ERBB2, ERBB3, ERK1/2, FOXO1/3a,
GSK3α/β, NF-κB, P, p38, p53, p70S6K, PRAS, PTEN, RB, S, RPS6, T, TSC2’. An
activating edge in the consensus network, as described in Additional file 3,
means that the sampled activating edges have a significantly higher
confidence value than the inhibiting ones. As self-loops and ingoing edges
to the drug or growth factor nodes were not allowed during inference, the
respective confidences are zero.
Additional file 5: Boolean interaction rules for the components of the
short- and long-term signalling networks. The tables contain the rules
that arose from network reconstructions based on short- and long-term
RPPA data of BT474, HCC1954 and SKBR3. The three drug names erlotinib,
trastuzumab and pertuzumab are abbreviated via their first letters. For the
long-term networks, the stimulus is denoted by S. Symbols are
interpretable in the following way: & ≡AND, ∨≡OR and ! ≡NOT.
Additional file 6: Reconstructed long-term signalling networks. The
figure displays the reconstructed long-term signalling networks for BT474,
HCC1954 and SKBR3. Target proteins are represented as rectangles with
stimulus and drugs coloured in red. The three drug names erlotinib,
trastuzumab and pertuzumab are abbreviated via their first letters.
Stimulation via full growth medium is denoted by S. Solid arrows denote
activating interactions while dashed ones represent inhibitions.
Abbreviations
DDEPN: Dynamic deterministic effects propagation networks; EGF: Epidermal
growth factor; EGFR: Epidermal growth factor receptor; GEO: Gene expression
omnibus; HER2: Human epidermal growth factor receptor 2; HMM: Hidden
Markov model; HRG: Heregulin; MAPK: Mitogen-activated protein kinase;
MCMC: Markov chain Monte Carlo; NIR: Near infrared; PI3K: Phosphoinositide
3-kinase; RPPA: Reverse phase protein array; RTK: Receptor tyrosine kinase.
Competing interests
The authors declare that they have no competing interests.
Authors’ contributions
FH performed the RPPA measurements under supervision of UK and was
mainly involved in target selection for the modelling approach. JS was
involved in discussing the conducted RPPA experiments. CB and TB
developed the applied network reconstruction algorithm and participated in
planning the modelling procedure. TB and SvdH initiated the simulation study
concepts. SvdH carried out the literature research, network reconstructions,
perturbation simulations followed by associated analyses, and finally drafting
the manuscript. All authors edited, read and approved the final manuscript.
Acknowledgements
Pertuzumab, trastuzumab and erlotinib were provided by Roche Diagnostics
GmbH, Penzberg, Germany. This work was supported by a grant from the
German Federal Ministry of Education and Research (BMBF) within the Medical
Systems Biology programme BreastSys.
We also acknowledge support by the Open Access Publication Funds of the
Göttingen University.
Author details
1Statistical Bioinformatics, Department of Medical Statistics, University Medical
Center Göttingen, Humboldtallee 32, 37073 Göttingen, Germany. 2TRON -
Translational Oncology at the University Medical Center Mainz,
Langenbeckstraße 1, 55131 Mainz, Germany. 3Science for Life Laboratory,
School of Biotechnology, KTH - Royal Institute of Technology, Box 1031, 17121
Solna, Sweden. 4Division of Molecular Genome Analysis, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg,
Germany.
Received: 20 December 2013 Accepted: 10 June 2014
Published: 25 June 2014
References
1.
Hill SM, Lu Y, Molina J, Heiser LM, Spellman PT, Speed TP, Gray JW, Mills
GB, Mukherjee S: Bayesian inference of signaling network topology in
a cancer cell line. Bioinformatics (Oxford, England) 2012,
28(21):2804–2810. PMID: 22923301.
2.
Park Y, Bader JS: How networks change with time. Bioinformatics
(Oxford, England) 2012, 28(12):40–48. PMID: 22689777.
3.
Roukos DH: Trastuzumab and beyond: sequencing cancer genomes
and predicting molecular networks. Pharmacogenom J 2011,
11(2):81–92. PMID: 20975737.
4.
Oda K, Matsuoka Y, Funahashi A, Kitano H: A comprehensive pathway
map of epidermal growth factor receptor signaling. Mol Syst Biol
2005, 1:2005.0010. PMID: 16729045.
5.
Feiglin A, Hacohen A, Sarusi A, Fisher J, Unger R, Ofran Y: Static network
structure can be used to model the phenotypic effects of
perturbations in regulatory networks. Bioinformatics (Oxford, England)
2012, 28(21):2811–2818. PMID: 22923292.
6.
Bender C, Heyde SV, Henjes F, Wiemann S, Korf U, Beissbarth T: Inferring
signalling networks from longitudinal data using sampling based
approaches in the r-package ‘ddepn’. BMC Bioinformatics 2011, 12:291.
PMID: 21771315.
7.
Penfold CA, Buchanan-Wollaston V, Denby KJ, Wild DL: Nonparametric
bayesian inference for perturbed and orthologous gene regulatory
networks. Bioinformatics 2012, 28(12):233–241. PMID: 22689766.
8.
Wagner JP, Wolf-Yadlin A, Sevecka M, Grenier JK, Root DE, Lauffenburger
DA, MacBeath G: Receptor tyrosine kinases fall into distinct classes
based on their inferred signaling networks. Sci Signaling 2013,
6(284):58.
9.
Chen WW, Schoeberl B, Jasper PJ, Niepel M, Nielsen UB, Lauffenburger
DA, Sorger PK: Input-output behavior of ErbB signaling pathways as
revealed by a mass action model trained against dynamic data. Mol
Syst Biol 2009, 5:PMID: 19156131.
10. Hatakeyama M, Kimura S, Naka T, Kawasaki T, Yumoto N, Ichikawa M, Kim
J-H, Saito K, Saeki M, Shirouzu M, Yokoyama S, Konagaya A: A
computational model on the modulation of mitogen-activated
protein kinase (MAPK) and akt pathways in heregulin-induced ErbB
signalling. Biochem J 2003, 373(Pt 2):451–463. PMID: 12691603.
11. Jones RB, Gordus A, Krall JA, MacBeath G: A quantitative protein
interaction network for the ErbB receptors using protein
microarrays. Nature 2006, 439(7073):168–174. PMID: 16273093.
12. Schoeberl B, Eichler-Jonsson C, Gilles ED, Müller G: Computational
modeling of the dynamics of the MAP kinase cascade activated by
surface and internalized EGF receptors. Nat Biotechnol 2002,
20(4):370–375. PMID: 11923843.
13. Sahin O, FrÃ˝uhlich H, LÃ˝ubke C, Korf U, Burmester S, Majety M, Mattern J,
Schupp I, Chaouiya C, Thieffry D, Poustka A, Wiemann S, Beissbarth T, Arlt
D: Modeling ERBB receptor-regulated G1/S transition to find novel
targets for de novo trastuzumab resistance. BMC Syst Biol 2009, 3:1.
PMID: 19118495.
14. Samaga R, Saez-Rodriguez J, Alexopoulos LG, Sorger PK, Klamt S: The
logic of EGFR/ErbB signaling: theoretical properties and analysis of
high-throughput data. PLoS Comput Biol 2009, 5(8):1000438. PMID:
19662154.
15. Wang R-S, Saadatpour A, Albert R: Boolean modeling in systems
biology: an overview of methodology and applications. Phys Biol
2012, 9(5):055001.
16. McDermott JE, Wang J, Mitchell H, Webb-Robertson B-J, Hafen R, Ramey J,
Rodland KD: Challenges in biomarker discovery: Combining expert
insights with statistical analysis of complex omics data. Expert Opin
Med Diag 2013, 7(1):37–51. PMID: 23335946.
17. Eduati F, De Las Rivas J, Di Camillo B, Toffolo G, Saez-Rodriguez J:
Integrating literature-constrained and data-driven inference of
signalling networks. Bioinformatics (Oxford, England) 2012,
28(18):2311–2317. PMID: 22734019.
18. Terfve C, Cokelaer T, Henriques D, MacNamara A, Goncalves E, Morris MK,
van Iersel M, Lauffenburger DA, Saez-Rodriguez J: CellNOptR: a flexible
toolkit to train protein signaling networks to data using multiple
logic formalisms. BMC Syst Biol 2012, 6:133. PMID: 23079107.
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 20 of 21
http://www.biomedcentral.com/1752-0509/8/75
19. Albert I, Thakar J, Li S, Zhang R, Albert R: Boolean network simulations
for life scientists. Source Code Biol Med 2008, 3:16. PMID: 19014577.
20. R Core Team: R: A Language and Environment for Statistical Computing.
Vienna, Austria: R Foundation for Statistical Computing; 2012. R
Foundation for Statistical Computing. ISBN 3-900051-07-0.
http://www.r-project.org.
21. Müssel C, Hopfensitz M, Kestler HA: BoolNet–an r package for
generation, reconstruction and analysis of boolean networks.
Bioinformatics (Oxford, England) 2010, 26(10):PMID: 20378558.
22. Samaga R, Klamt S: Modeling approaches for qualitative and semi-
quantitative analysis of cellular signaling networks. Cell Commun
Signaling: CCS 2013, 11(1):43. PMID: 23803171 PMCID: PMC3698152.
23. Gonzalez AG, Naldi A, Sánchez L, Thieffry D, Chaouiya C: GINsim: a
software suite for the qualitative modelling, simulation and analysis
of regulatory networks. Bio Syst 2006, 84(2):91–100. PMID: 16434137.
24. Di Cara A, Garg A, De Micheli G, Xenarios I, Mendoza L: Dynamic
simulation of regulatory networks using SQUAD. BMC Bioinformatics
2007, 8:462. PMID: 18039375 PMCID: PMC2238325.
25. Helikar T, Rogers JA: ChemChains: a platform for simulation and
analysis of biochemical networks aimed to laboratory scientists.
BMC Syst Biol 2009, 3:58. PMID: 19500393 PMCID: PMC2705353.
26. Krumsiek J, Pösterl S, Wittmann DM, Theis FJ: Odefy–from discrete to
continuous models. BMC Bioinformatics 2010, 11:233. PMID: 20459647
PMCID: PMC2873544.
27. Ferlay J, Shin H-R, Bray F, Forman D, Mathers C, Parkin DM: Estimates of
worldwide burden of cancer in 2008: Globocan 2008. Int J Cancer
2010, 127(12):2893–2917.
28. Henjes F, Bender C, Heyde SVD, Braun L, Mannsperger HA, Schmidt C,
Wiemann S, Hasmann M, Aulmann S, Beissbarth T, Korf U: Strong EGFR
signaling in cell line models of ERBB2-amplified breast cancer
attenuates response towards ERBB2-targeting drugs. Oncogenesis
2012, 1(7):16.
29. Olayioye MA, Neve RM, Lane HA, Hynes NE: The ErbB signaling
network: receptor heterodimerization in development and cancer.
EMBO J 2000, 19(13):PMID: 10880430.
30. Heil J, Gondos A, Rauch G, Marmé F, Rom J, Golatta M, Junkermann H,
Sinn P, Aulmann S, Debus J, Hof H, Schütz F, Brenner H, Sohn C,
Schneeweiss A: Outcome analysis of patients with primary breast
cancer initially treated at a certified academic breast unit. Breast
(Edinburgh, Scotland) 2012, 21(3):303–308. PMID: 22310244.
31. Jelovac D, Wolff AC: The adjuvant treatment of HER2-positive breast
cancer. Curr Treat Options Oncol 2012, 13(2):230–239. PMID: 22410709.
32. Park JW, Neve RM, Szollosi J, Benz CC: Unraveling the biologic and
clinical complexities of HER2. Clin Breast Cancer 2008, 8(5):392–401.
PMID: 18952552.
33. Tinoco G, Warsch S, Glück S, Avancha K, Montero AJ: Treating breast
cancer in the 21st century: emerging biological therapies. J Cancer
2013, 4(2):117–132. PMID: 23386910.
34. Heyde Svd, Beissbarth T: A new analysis approach of epidermal
growth factor receptor pathway activation patterns provides
insights into cetuximab resistance mechanisms in head and neck
cancer. BMC Medicine 2012, 10(1):43. PMID: 22548923.
35. Motoyama AB, Hynes NE, Lane HA: The efficacy of ErbB
receptor-targeted anticancer therapeutics is influenced by the
availability of epidermal growth factor-related peptides. Cancer Res
2002, 62(11):3151–3158.
36. Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, Peng J, Lin E,
Wang Y, Sosman J, Ribas A, Li J, Moffat J, Sutherlin DP, Koeppen H,
Merchant M, Neve R, Settleman J: Widespread potential for
growth-factor-driven resistance to anticancer kinase inhibitors.
Nature 2012, 487(7408):505–509. PMID: 22763448.
37. Gallardo A, Lerma E, Escuin D, Tibau A, Muñoz J, Ojeda B, Barnadas A,
Adrover E, Sánchez-Tejada L, Giner D, Ortiz-Martínez F, Peiró G: Increased
signalling of EGFR and IGF1R, and deregulation of PTEN/PI3K/Akt
pathway are related with trastuzumab resistance in HER2 breast
carcinomas. Br J Cancer 2012, 106(8):1367–1373. PMID: 22454081.
38. Wang L, Zhang Q, Zhang J, Sun S, Guo H, Jia Z, Wang B, Shao Z, Wang Z,
Hu X: PI3K pathway activation results in low efficacy of both
trastuzumab and lapatinib. BMC Cancer 2011, 11:248. PMID: 21676217.
39. Diermeier S, Horvà ˛ath G, Knuechel-Clarke R, Hofstaedter F, Söllosi J,
Brockhoff G: Epidermal growth factor receptor coexpression
modulates susceptibility to herceptin in HER2/neu overexpressing
breast cancer cells via specific erbB-receptor interaction and
activation. Exp Cell Res 2005, 304(2):604–619. PMID: 15748904.
40. Pallis AG, Syrigos KN: Epidermal growth factor receptor tyrosine
kinase inhibitors in the treatment of NSCLC. Lung cancer (Amsterdam,
Netherlands) (2013). PMID: 23384674.
41. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ,
Murawa P, Walde D, Wolff RA, Campos D, Lim R, Ding K, Clark G,
Voskoglou-Nomikos T, Ptasynski M, Parulekar W, National Cancer, Institute
of Canada Clinical Trials Group: Erlotinib plus gemcitabine compared
with gemcitabine alone in patients with advanced pancreatic
cancer: a phase III trial of the national cancer institute of canada
clinical trials group. J Clin Oncol 2007, 25(15):1960–1966. PMID:
17452677.
42. Loebke C, Sueltmann H, Schmidt C, Henjes F, Wiemann S, Poustka A, Korf
U: Infrared-based protein detection arrays for quantitative
proteomics. PROTEOMICS 2007, 7(4):558–564.
43. Kataoka Y, Mukohara T, Shimada H, Saijo N, Hirai M, Minami H:
Association between gain-of-function mutations in PIK3CA and
resistance to HER2-targeted agents in HER2-amplified breast cancer
cell lines. Ann Oncol 2010, 21(2):255–262. PMID: 19633047.
44. Nahta R, Yuan LXH, Zhang B, Kobayashi R, Esteva FJ: Insulin-like growth
factor-i receptor/human epidermal growth factor receptor 2
heterodimerization contributes to trastuzumab resistance of breast
cancer cells. Cancer Res 2005, 65(23):11118–11128. PMID: 16322262.
45. Hornbeck PV, Kornhauser JM, Tkachev S, Zhang B, Skrzypek E, Murray B,
Latham V, Sullivan M: PhosphoSitePlus: a comprehensive resource for
investigating the structure and function of experimentally
determined post-translational modifications in man and mouse.
Nucleic Acids Res 2011, 40(D1):261–270.
46. FrÃ˝uhlich H, Sahin O, Arlt D, Bender C, Beissbarth T: Deterministic effects
propagation networks for reconstructing protein signaling
networks from multiple interventions. BMC Bioinformatics 2009,
10:322. PMID: 19814779.
47. Bender C, Henjes F, Fröhlich H, Wiemann S, Korf U, Beissbarth T: Dynamic
deterministic effects propagation networks: learning signalling
pathways from longitudinal protein array data. Bioinformatics (Oxford,
England) 2010, 26(18):596–602. PMID: 20823327.
48. Almudevar A, McCall MN, McMurray H, Land H: Fitting boolean
networks from steady state perturbation data. Stat Appl Genet Mol Biol
2011, 10(1):1–40.
49. Brooks SP, Gelman A: General methods for monitoring convergence
of iterative simulations. J Comput Graph Stat 1998, 7(4):434–455.
50. Benjamini Y, Hochberg Y: Controlling the false discovery rate: A
practical and powerful approach to multiple testing. J R Stat Soc Series
B (Methodological) 1995, 57(1):289–300. ArticleType: research-article/Full
publication date: 1995/Copyright ©1995 Royal Statistical Society.
51. Bo Na Ki, D, Odenbrett MR, Wijs A, Ligtenberg W, Hilbers P: Efficient
reconstruction of biological networks via transitive reduction on
general purpose graphics processors. BMC Bioinformatics 2012,
13(1):PMID: 23110660.
52. Mikalsen T, Gerits N, Moens U: Inhibitors of signal transduction protein
kinases as targets for cancer therapy. In M Raafat El-Gewely (ed.)
Biotechnology Annual Review. Volume 12: Elsevier:153–223. ISBN:
1387-2656 2006. http://www.sciencedirect.com/science/article/pii/
S1387265606120062.
53. Dienstmann R, De Dosso S, Felip E, Tabernero J: Drug development to
overcome resistance to EGFR inhibitors in lung and colorectal
cancer. Mol Oncol 2012, 6(1):15–26.
54. Esteva FJ, Pusztai L: Optimizing outcomes in HER2-positive breast
cancer: the molecular rationale. Oncology (Williston Park, N.Y.) 2005,
19(13 Suppl 5):5–16. PMID: 19364051.
55. Sato S, Fujita N, Tsuruo T: Involvement of 3-phosphoinositide-
dependent protein kinase-1 in the MEK/MAPK signal transduction
pathway. J Biol Chem 2004, 279(32):33759–33767. PMID: 15175348.
56. Maurer M, Su T, Saal LH, Koujak S, Hopkins BD, Barkley CR, Wu J, Nandula S,
Dutta B, Xie Y, Chin YR, Kim D-I, Ferris JS, Gruvberger-Saal SK, Laakso M,
Wang X, Memeo L, Rojtman A, Matos T, Yu JS, Cordon-Cardo C, Isola J,
Terry MB, Toker A, Mills GB, Zhao JJ, Murty VVVS, Hibshoosh H, Parsons R:
3-phosphoinositide-dependent kinase 1 potentiates upstream
lesions on the phosphatidylinositol 3-kinase pathway in breast
carcinoma. Cancer Res 2009, 69(15):6299–6306. PMID: 19602588.
von der Heyde et al. BMC Systems Biology 2014, 8:75
Page 21 of 21
http://www.biomedcentral.com/1752-0509/8/75
57. Tseng P-H, Wang Y-C, Weng S-C, Weng J-R, Chen C-S, Brueggemeier RW,
Shapiro CL, Chen C-Y, Dunn SE, Pollak M, Chen C-S: Overcoming
trastuzumab resistance in HER2-overexpressing breast cancer cells
by using a novel celecoxib-derived phosphoinositide-dependent
kinase-1 inhibitor. Mol Pharmacol 2006, 70(5):PMID: 16887935.
58. Vega F, Medeiros LJ, Leventaki V, Atwell C, Cho-Vega JH, Tian L, Claret F-X,
Rassidakis GZ: Activation of mammalian target of rapamycin
signaling pathway contributes to tumor cell survival in anaplastic
lymphoma kinase-positive anaplastic large cell lymphoma. Cancer
Res 2006, 66(13):6589–6597. PMID: 16818631.
59. Frödin M, Jensen CJ, Merienne K, Gammeltoft S: A phosphoserine-
regulated docking site in the protein kinase RSK2 that recruits and
activates PDK1. EMBO J 2000, 19(12):2924–2934. PMID: 10856237.
60. Klos KS, Wyszomierski SL, Sun M, Tan M, Zhou X, Li P, Yang W, Yin G,
Hittelman WN, Yu D: ErbB2 increases vascular endothelial growth
factor protein synthesis via activation of mammalian target of
rapamycin/p70S6K leading to increased angiogenesis and
spontaneous metastasis of human breast cancer cells. Cancer Res
2006, 66(4):2028–2037. PMID: 16489002.
61. Schaefer G, Shao L, Totpal K, Akita RW: Erlotinib directly inhibits HER2
kinase activation and downstream signaling events in intact cells
lacking epidermal growth factor receptor expression. Cancer Res
2007, 67(3):1228–1238. PMID: 17283159.
62. Zhong Q, Simonis N, Li Q-R, Charloteaux B, Heuze F, Klitgord N, Tam S, Yu
H, Venkatesan K, Mou D, Swearingen V, Yildirim MA, Yan H, Dricot A, Szeto
D, Lin C, Hao T, Fan C, Milstein S, Dupuy D, Brasseur R, Hill DE, Cusick ME,
Vidal M: Edgetic perturbation models of human inherited disorders.
Mol Syst Biol 2009, 5:321. PMID: 19888216.
63. McCurrach ME, Connor TM, Knudson CM, Korsmeyer SJ, Lowe SW:
bax-deficiency promotes drug resistance and oncogenic
transformation by attenuating p53-dependent apoptosis. Proc Nat
Acad Sci USA 1997, 94(6):2345–2349. PMID: 9122197.
64. Sherr CJ, McCormick F: The RB and p53 pathways in cancer. Cancer Cell
2002, 2(2):103–112. PMID: 12204530.
65. Sithanandam G, Anderson LM: The ERBB3 receptor in cancer and
cancer gene therapy. Cancer Gene Therapy 2008, 15(7):413–448. PMID:
18404164.
66. Lynch DK, Daly RJ: PKB-mediated negative feedback tightly regulates
mitogenic signalling via gab2. EMBO J 2002, 21(1-2):72–82. PMID:
11782427.
67. Chakrabarty A, Sánchez V, Kuba MG, Rinehart C, Arteaga CL: Feedback
upregulation of HER3 (ErbB3) expression and activity attenuates
antitumor effect of PI3K inhibitors. Proc Nat Acad Sci USA 2012,
109(8):2718–2723. PMID: 21368164.
68. Abrieu A, Dorée M, Fisher D: The interplay between cyclin-b-cdc2
kinase (MPF) and MAP kinase during maturation of oocytes. J Cell Sci
2001, 114(Pt 2):257–267. PMID: 11148128.
69. Jirmanova L, Afanassieff M, Gobert-Gosse S, Markossian S, Savatier P:
Differential contributions of ERK and PI3-kinase to the regulation of
cyclin d1 expression and to the control of the G1/S transition in
mouse embryonic stem cells. Oncogene 2002, 21(36):5515–5528. PMID:
12165850.
70. Searle JS, Li B, Du W: Targeting rb mutant cancers by inactivating
TSC2. Oncotarget 2010, 1(3):228–232. PMID: 20706560.
71. Liu H, Radisky DC, Nelson CM, Zhang H, Fata JE, Roth RA, Bissell MJ:
Mechanism of akt1 inhibition of breast cancer cell invasion reveals a
protumorigenic role for TSC2. Proc Nat Acad Sci USA 2006,
103(11):4134–4139. PMID: 16537497.
doi:10.1186/1752-0509-8-75
Cite this article as: von der Heyde et al.: Boolean ErbB network
reconstructions and perturbation simulations reveal individual drug
response in different breast cancer cell lines. BMC Systems Biology 2014 8:75.
Submit your next manuscript to BioMed Central
and take full advantage of:
• Convenient online submission
• Thorough peer review
• No space constraints or color figure charges
• Immediate publication on acceptance
• Inclusion in PubMed, CAS, Scopus and Google Scholar
• Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
|
24970389
|
p70S6K = ( ( ( ( ERK1_2 ) AND NOT ( TSC2 ) ) AND NOT ( Nfkb ) ) AND NOT ( PRAS ) ) OR ( ( ( ( p70S6K ) AND NOT ( TSC2 ) ) AND NOT ( Nfkb ) ) AND NOT ( PRAS ) )
Nfkb = ( Nfkb )
FoxO1_3a = ( ( FoxO1_3a ) AND NOT ( AKT ) )
AKT = ( ( ERBB3 ) AND NOT ( PTEN ) ) OR ( ( AKT ) AND NOT ( PTEN ) ) OR ( ( ERBB1 ) AND NOT ( PTEN ) ) OR ( ( ERBB2 ) AND NOT ( PTEN ) )
BAX = ( BAX )
p53 = ( CyclinB1 ) OR ( PTEN ) OR ( RB ) OR ( p38 ) OR ( stimulus ) OR ( p53 )
CyclinB1 = ( ( ERBB3 ) AND NOT ( p53 ) ) OR ( ( CyclinB1 ) AND NOT ( p53 ) ) OR ( ( ERBB1 ) AND NOT ( p53 ) )
ERBB1 = ( ( ( ERBB1 ) AND NOT ( pertuzumab ) ) AND NOT ( erlotinib ) ) OR ( ( ( stimulus ) AND NOT ( pertuzumab ) ) AND NOT ( erlotinib ) )
cRAF = ( ( cRAF ) AND NOT ( ERK1_2 ) ) OR ( ( ERBB1 ) AND NOT ( ERK1_2 ) ) OR ( ( ERBB2 ) AND NOT ( ERK1_2 ) )
p38 = ( AKT ) OR ( p38 )
RPS6 = ( p70S6K ) OR ( RPS6 )
ERBB2 = ( ( ( ( stimulus ) AND NOT ( pertuzumab ) ) AND NOT ( trastuzumab ) ) AND NOT ( erlotinib ) ) OR ( ( ( ( ERBB2 ) AND NOT ( pertuzumab ) ) AND NOT ( trastuzumab ) ) AND NOT ( erlotinib ) )
RB = ( ( RB ) AND NOT ( CyclinD1 ) ) OR ( ( Nfkb ) AND NOT ( CyclinD1 ) )
ERBB3 = ( ( ( ( ERBB3 ) AND NOT ( PTEN ) ) AND NOT ( erlotinib ) ) AND NOT ( pertuzumab ) ) OR ( ( ( ( RPS6 ) AND NOT ( PTEN ) ) AND NOT ( erlotinib ) ) AND NOT ( pertuzumab ) ) OR ( ( ( ( stimulus ) AND NOT ( PTEN ) ) AND NOT ( erlotinib ) ) AND NOT ( pertuzumab ) )
GSK3a_b = ( CyclinD1 ) OR ( GSK3a_b ) OR ( p53 )
cJUN = ( ( cJUN ) AND NOT ( GSK3a_b ) )
CyclinD1 = ( ( AKT ) AND NOT ( GSK3a_b ) ) OR ( ( CyclinD1 ) AND NOT ( GSK3a_b ) ) OR ( ( ERK1_2 ) AND NOT ( GSK3a_b ) ) OR ( ( RPS6 ) AND NOT ( GSK3a_b ) )
TSC2 = ( ( ( ( TSC2 ) AND NOT ( AKT ) ) AND NOT ( ERK1_2 ) ) AND NOT ( GSK3a_b ) )
PRAS = ( ( PRAS ) AND NOT ( AKT ) )
ERK1_2 = ( ERK1_2 ) OR ( cRAF )
PTEN = ( ( PTEN ) AND NOT ( GSK3a_b ) )
|
ORIGINAL RESEARCH ARTICLE
published: 05 December 2014
doi: 10.3389/fimmu.2014.00599
Design, assessment, and in vivo evaluation of a
computational model illustrating the role of CAV1 in CD4+
T-lymphocytes
Brittany D. Conroy 1†,Tyler A. Herek 1†,Timothy D. Shew 1†, Matthew Latner 1, Joshua J. Larson1,
Laura Allen1, Paul H. Davis 1,Tomáš Helikar 2 and Christine E. Cutucache1*
1 Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
2 Department of Biochemistry, University of Nebraska at Lincoln, Lincoln, NE, USA
Edited by:
Sergio Quezada, University College
London Cancer Institute, UK
Reviewed by:
Carlos Alfaro, Clínica Universidad de
Navarra, Spain
Haidong Dong, Mayo Clinic, USA
*Correspondence:
Christine E. Cutucache, Department
of Biology, University of Nebraska at
Omaha, Allwine Hall 413, 6001 Dodge
Street, Omaha, NE 68182, USA
e-mail: ccutucache@unomaha.edu
†Co-first authors of this manuscript
Caveolin-1 (CAV1) is a vital scaffold protein heterogeneously expressed in both healthy and
malignant tissue. We focus on the role of CAV1 when overexpressed in T-cell leukemia.
Previously, we have shown that CAV1 is involved in cell-to-cell communication, cellular
proliferation, and immune synapse formation; however, the molecular mechanisms have
not been elucidated. We hypothesize that the role of CAV1 in immune synapse formation
contributes to immune regulation during leukemic progression, thereby warranting stud-
ies of the role of CAV1 in CD4+ T-cells in relation to antigen-presenting cells. To address
this need, we developed a computational model of a CD4+ immune effector T-cell to
mimic cellular dynamics and molecular signaling under healthy and immunocompromised
conditions (i.e., leukemic conditions). Using the Cell Collective computational modeling
software, the CD4+ T-cell model was constructed and simulated under CAV1+/+, CAV1+/−,
and CAV1−/−conditions to produce a hypothetical immune response. This model allowed
us to predict and examine the heterogeneous effects and mechanisms of CAV1 in silico.
Experimental results indicate a signature of molecules involved in cellular proliferation, cell
survival, and cytoskeletal rearrangement that were highly affected by CAV1 knock out.
With this comprehensive model of a CD4+ T-cell, we then validated in vivo protein expres-
sion levels. Based on this study, we modeled a CD4+ T-cell, manipulated gene expression
in immunocompromised versus competent settings, validated these manipulations in an
in vivo murine model, and corroborated acute T-cell leukemia gene expression profiles in
human beings. Moreover, we can model an immunocompetent versus an immunocom-
promised microenvironment to better understand how signaling is regulated in patients
with leukemia.
Keywords: caveolin-1, CD4+ T-lymphocyte, the cell collective, adult T-cell leukemia, immunosuppression,
immunotherapy, computational biology, logical models
INTRODUCTION
Caveolae are cave-like invaginations comprised mostly of the pro-
tein caveolin-1 (CAV1). In addition to the traditional roles of
CAV1 in endocytosis, CAV1 has been implicated in processes
ranging from signal transduction (1, 2), to both oncogenesis (3–
5), and tumor suppression (6–8). Recently, three new roles for
CAV1 emerged, including regulating immune synapse formation,
T-cell receptor (TCR) activation, and mediating actin polymer-
ization (9–11). Caveolin-1 knockout studies show an attenuated
immunesynapseformationasobservedbydecreasedF-actinstain-
ing and dysregulation of RAC1 and ARP2/3 pathways (9). When
T-cells engage with antigen-presenting cells (APCs), decreased
TCR-dependent T-cell proliferation isobserved when CAV1ispro-
hibited from interacting with CD26 (12). Downstream signaling
pathways affected by CAV1 knockdown include the organization
of the KSR1 mediated Raf/MEK/ERK signal cascade (13) and
ZAP70, p56lck, and TCRζ phosphorylation (14). This mechanism
has been shown to be distinct from CD3/CD28 stimulation (15),
where no proliferation defects were observed in Cav1−/−T-cells.
CAV1 acts as a scaffolding molecule thereby likely contributing
to diverse events in the cell through CAV1-mediated recruit-
ment of signaling complexes to the plasma membrane. More-
over, T-cell activation through the TCR and competent immune
synapse formation are necessary for a healthy immune response.
Misregulation of these processes can lead to deleterious effects,
including cancer progression and a phenotype known as tumor-
induced immunosuppression (16, 17). As CD4+ T-cells are vital
for proper adaptive immune function, and CAV1 plays a role in
immune synapse formation, we chose to further investigate the
CAV1-mediated pathways in a CD4+ T-cell.
To better understand the intricate biology of CAV1 signaling
in CD4+ T-cells, the development of a comprehensive in silico
model is warranted. Through such a model, further identification
of molecules associated with CAV1 signaling can occur.
www.frontiersin.org
December 2014 | Volume 5 | Article 599 | 1
Conroy et al.
Role of CAV1 in CD4+ T-lymphocytes
Importantly, such a model allows for real-time simulations
using computer software in an effort to identify specific mech-
anisms in the cell. In order to generate such a comprehensive and
dynamic model, a systems biology approach is required (18). This
approach provides a potentially greater understanding of the com-
plex cellular functions that occur in living systems, allowing the
use of computer models to conduct thousands of virtual experi-
ments as well as make methodical predictions regarding proteins
of interest (18–21).
Herein, we describe the construction and validation of a fully
functional in silico CD4+ T-cell model using the Cell Collec-
tive, a web-based, open-source dynamic modeling platform that
allows scientists to construct computational models in a non-
mathematical fashion (20,22). From this in silico starting position,
comprehensive simulations were performed, allowing for predic-
tions and hypotheses to be drawn for further in vitro/in vivo
experimentation. To our knowledge,this is the first time a dynamic
model of a CD4+ T-cell has been created to observe the down-
stream effects of CAV1+/+ (wild type), CAV1+/−(heterozygous),
and CAV1−/−(knock down) upon cell signaling and intracel-
lular networks as validated by in silico simulations and in vivo
investigations.
MATERIALS AND METHODS
COMPUTATIONAL MODEL CONSTRUCTION WITH CELL COLLECTIVE
The presented model was constructed using Cell Collective –
a collaborative and interactive platform for modeling biologi-
cal/biochemical systems (20, 22). The mathematical framework
behind Cell Collective is based on a common qualitative (discrete)
modeling technique where the regulatory mechanism of each node
isdescribedwithalogicalfunction[formorecomprehensiveinfor-
mation on logical modeling, see Ref. (23, 24)]. Cell Collective
allows users to construct and simulate large-scale computational
models of various biological processes based on qualitative inter-
action information extracted from previously published literature.
The initial version of the model was structured after the previously
published models (25, 26). The individual components and local
interactions in the presented final model were retrieved manually
from published literature. The model was subsequently validated
against well-known experimentally demonstrated T-cell dynam-
ics (see Model Validation), as well as new experiments presented
in this paper. The Cell Collective’s Knowledge Base was used to
catalog and annotate every interaction and regulatory mechanism
(e.g., tyrosine phosphorylation on Y316) as mined from the pri-
mary literature. The model is freely available for simulations and
furthercontributionsbyothers directlyintheplatform. Themodel
can be also downloaded in the SBML format (24) to be used within
other software tools.
MODEL VALIDATION
Themodel was constructed usinglocal (e.g.,protein–protein inter-
action) information from the primary literature. In other words,
during the construction phase of the model, there was no attempt
to determine the local interactions based on any other larger phe-
notypes or phenomena. However, after the model was completed,
verification of the accuracy of the model involved testing it for
the ability to reproduce complex input–output phenomena that
have been observed in the laboratory. To do this, the T-cell model
was simulated under a multitude of cellular conditions and ana-
lyzed in terms of input–output dose–response curves to determine
whether the model behaves as expected [Figure 2; Ref. (27–33)],
including various downstream effects as a result of activation of
the TCR,G-protein-coupled receptor,cytokine,and integrin path-
ways. A total of 20 phenomena were used for the validation phase
(data not shown).
IN SILICO SIMULATIONS
The Cell Collective platform was utilized to perform all sim-
ulations for the CD4+ T-cell model. Virtual extracellular envi-
ronments, composed of 20 CD4+ T-cell stimuli, were optimized
for each in silico experiment based on immunocompetent versus
immunocompromised (diseased) settings [Table 1; Ref. (9, 13, 30,
Table 1 | A summary of the experimental conditions simulated.
External
stimulus
Tissue
(WT)
Tissue
(Disease A)
Tissue
(Disease B)
Alpha_13L
Med
High
High
GalphaS_L
Med
High
High
APC
Med
High
High
CGC
Med
Med-High
Med-High
ECM
High
High
High
GP130
0
0
0
IFNB
Med
Med
Med
IFNG
Med
Med
Med
IFNGR1
Med
Med
Med
IFNGR2
Med
Med
Med
IL10
Med
High
0–100
IL10RA
Med
High
High
IL10RB
Med
Med
Med
IL12
Med
High
High
IL15
Med
High
High
IL15RA
Med
High
High
IL18
Med
High
High
IL21
Med
High
High
IL22
Med
High
High
IL23
Med
High
High
IL27
Med
High
High
IL27RA
Med
Med
Med
IL2
Med
High
High
IL2RB
Med
High
High
IL4
Low
Low
Low
IL6
Low
Low
Low
IL6RA
Low
Low
Low
IL9
Low
Low
Low
TGFB
Low
Low
Low
Specifically, these conditions included (1) a wild-type condition (i.e., healthy bio-
logical levels of cytokines), (2) an immunosuppressive disease condition (i.e.,
Disease A), and (3) a scenario with varying degrees of immunosuppression (i.e.,
Disease B) with CAV1+/+, CAV1+/−, and CAV1−/−.
Low, 0–20 activity level; Med, 21–60 activity level; High, 61–100 activity level
(Note that the activity levels do not directly correspond to concentrations, rather
the activity levels provide a semi-quantitative measure to describe the relative
activity of a particular component of the model.).
Frontiers in Immunology | Tumor Immunity
December 2014 | Volume 5 | Article 599 | 2
Conroy et al.
Role of CAV1 in CD4+ T-lymphocytes
FIGURE 1 | In silico modeling of a CD4+ T-cell. (A) Nodal representation of
CD4+ T-cell signaling pathways constructed using the Bio-Logic builder
inclusive within the Cell Collective. Linkages represent protein–protein,
protein–phosphorylation, and kinase interactions. (B) Osprey modeling of
predicted CAV1 protein–protein interactions and functions. Linkages are
categorized by function and centrality to CAV1. (C) Graphical depiction of
CAV1-associated interactions. Major pathway end-points include cell survival,
cytoskeletal rearrangement, and cellular proliferation.
31, 34, 35)]. For each experiment, these values were analyzed and
used to compare proteins most affected by CAV1+/+, CAV1+/−,
and CAV1−/−in an immunocompetent (i.e., WT) versus varying
degrees of immunosuppression (i.e., Diseases A versus B) condi-
tion. The model was simulated under hypothetical disease-causing
environments in order to observe the changes, if any, in CAV1 reg-
ulatory activity. Disease A mimics an immunosuppressive disease
condition, and disease B simulates varying degrees of immuno-
suppression, as controlled by varying IL-10 levels. Each experi-
ment consisted of 1,000 simulations, with different activity levels
randomly selected between 0 and 100 for the external stimuli
representing the extracellular environment. Each simulation con-
sisted of 800 iterations and activity level of the model species was
calculated over the last 300 iterations using methods previously
www.frontiersin.org
December 2014 | Volume 5 | Article 599 | 3
Conroy et al.
Role of CAV1 in CD4+ T-lymphocytes
FIGURE 2 |The Cell Collective accurately models complex cellular
phenomena. (A–F) Certification of Bio-Logic built local interactions
executing in accordance with primary literature findings. (A) Activation of
the mitogen-activated protein kinase (MAPK) pathway via APC stimulation
(27). (B) Positive relationship between filamentous actin polymerization in
response to stimulation with extracellular matrix (ECM) components (28).
(C) PI3-Kinase activation via binding of ligand to G protein-coupled
receptor, GαQ (29). (D) Activation of the MAPK pathway via
integrin-dependent ECM stimulation (9, 30). (E) Activation of the MAPK
pathway via stimulation with interleukin-2 (IL2) (31, 32). (F) Activation of
the small GTPase Cdc42 via binding of ligand to the G protein-coupled
receptor, Gα12/13 (33); these results not shown in the graphic. Each
dose–response curves appears to demonstrate a positive correlation with
the stimulus.
described (36). The aforementioned simulations were run under
six separate conditions including wild-type tissue, diseased tissue
(diseasesA and B),wild-type blood,and diseased blood (diseasesA
and B).Wild-type and diseased blood simulations are not included
duetoinconclusivedata. Eachexperimentalenvironmentwassim-
ulated under (1) healthy cellular conditions, (2) CAV1 knocked
out, (3) CAV1 activated 50% of the time (CAV1+/−) and (4) CAV1
activated at random levels between 0 and 100. (To be able to arti-
ficially control the activity levels of CAV1 under environments 3
and 4, an external species, “CAV1 Activator,” was built into the
model to activate CAV1 independently of the activity levels of its
direct upstream regulators).
MOUSE MAINTENANCE
Animals were housed in pathogen-free animal facilities, and all
experimental protocols were reviewed and approved per the
Institutional Animal Care and Use Committee at the University
of Nebraska Medical Center/University of Nebraska at Omaha
(IACUC# 13-056-08-EP). C57Bl/6J and B6.Cg-Cav1tm1Mls/J mice
were purchased from the Jack-son Laboratory (Bar Harbor,
ME, USA). Post-natal day 54 (±5 days) mice were used for all
experiments.
HISTOLOGICAL STAINING
Spleen and lymph node tissues were sectioned and stained at the
University of Nebraska Medical Center’s Tissue Science Facility.
Spleen and lymph node tissues were sectioned, preserved in 10%
formalin, and embedded onto slides in paraffin. Specifically, all
tissues were sectioned after at least 48 h in fixative and stained
with hematoxylin and eosin using standard protocol.
For immunohistochemical staining (IHC), tissues were depar-
rafinized in xylene for 3 min and rehydrated in decreasing concen-
trations of ethanol (100–50%). Antigen retrieval was performed
by boiling sections in a solution of Sodium Citrate with 0.05%
Tween-20. Blocking against non-specific binding and endoge-
nous peroxidases was performed by incubation with 5% bovine
serum albumin (BSA; Invitrogen) and 0.3% hydrogen perox-
ide, respectively. Primary antibody incubation was conducted for
90 min at room temperature in phosphate buffered saline (PBS;
DIBCO). The following antibodies were used: Rac1 (Merck KGaA,
Darmstadt, Germany); CD28 (BD Pharmingen); GATA3 (BD
Pharmingen); CD26 (Abcam); BCL10 (Cell Applications, Inc.).
Horse radish peroxidase-conjugated secondary antibodies (Cell
Signaling Technology; BD Pharmingen; Abcam) were incubated
for 1 h at room temperature. Slides were developed with a working
solution 3,3-diaminobenzidine for 10 min at room temperature,
followedbyrinsing withdistilledwaterand mountingthecoverslip
with Permount (Thermo Fisher Scientific).
GENE EXPRESSION PROFILING
Microarray data were downloaded from the Gene Expression
Omnibus, accession number GSE55851 (37). Data contained
whole genome expression profiling of CD4+ T-cells, sorted based
on a CADM1/CD7 phenotype. Samples were collected from
Frontiers in Immunology | Tumor Immunity
December 2014 | Volume 5 | Article 599 | 4
Conroy et al.
Role of CAV1 in CD4+ T-lymphocytes
FIGURE 3 | In silico predictions for translation into in vitro/in vivo
experimentation. Following 1,000 iterations of simulation as described in
Table 1, the most affected proteins (either up or downregulated) were
compiled by the Cell Collective and ranked based on activity% ON.
Specifically, those described were the top 15 most differentially expressed
molecules in the (A) CAV1+/+, (B) Cav1+/−, (C) CAV1−/−genotype. These
proteins were selected for further investigation with in vitro/in vivo
verification.
patients diagnosed with Adult T-cell leukemia-lymphoma (ATL)
subtypes: asymptomatic (n = 2), smoldering (n = 2), chronic
(n = 1), acute (n = 2), and healthy controls (n = 3). A mini-
mum of two samples were taken from each patient for microar-
ray analyses. Molecules of interest, as established utilizing the
Cell Collective, were selected within the microarray data and
analyzed by fold-change from normal controls between ATL
subtypes. Fold-change values were subjected to uncentered,
average-linkage correlation using Cluster 3.0, and Java Tree-
View as described previously (9). Further, Pearson regression
analyses were conducted to discern correlation among mole-
cules of interest in relation to CAV1 expression across ATL
subtypes.
RESULTS
IN SILICO MODELING OF A CD4+ T-CELL
The completed CD4+ T-cell model consists of 188 nodes
representing components of various signaling pathways and
corresponding protein-to-protein, protein-phosphorylation, and
kinase interactions (Figure 1A). These interactions correlate with
preliminary data generated using Osprey software used to model
CAV1 protein-to-protein interaction types (Figure 1B). Each
method of analysis (both the Cell Collective and Osprey) impli-
cates a role for CAV1 in phosphorylation, signal transduction,
transport, and cytoskeletal arrangement. The regulation of these
events by CAV1 is highly complex and dynamic, as illustrated in
Figure 1C.
To verify that the local interactions built into the model were
able to accurately mimic complex phenomena that have been
produced in the laboratory,20 validations were conducted via sim-
ulations in Cell Collective (six representative validations are shown
in Figures 2A–F). As an example, simulated TCR activation by an
APC leads to Erk activation,and the subsequent downstream effect
of cellular proliferation as represented by the literature (Figure 2A,
Figure S1 in Supplemental Material).
IDENTIFICATION OF MOST AFFECTED PROTEINS IN SILICO
Experiments were simulated using the in silico model (as described
in Table 1) to make rational predictions about how the sys-
tem would function in the laboratory. Following 1,000 iterations,
www.frontiersin.org
December 2014 | Volume 5 | Article 599 | 5
Conroy et al.
Role of CAV1 in CD4+ T-lymphocytes
we observed the protein products most affected by CAV1+/+,
CAV1+/−, and CAV1−/−in immounocompetent versus immuno-
compromised conditions (Figures 3A–C). In order to test the
validity of the model, we chose to investigate expression levels
of the proteins most affected in the knock down genotype as com-
pared to a wild-type system. Specifically proteins disregulated by
CAV1−/−across all three conditions (WT, Disease A, and Disease
B) included CD26, CARMA1, FYN, SHC1, SOS, SHP2, NOS2A,
BCL10, and GRB2 (Figure 3C). Based on findings from the model
as well as preliminary in vitro data, we hypothesized that CAV1
expression regulates Ras-related C3 botulinum toxin substrate 1
(RAC1), B-cell lymphoma/leukemia 10 (BCL10), GATA-binding
protein 3 (GATA3), CD26, and CD28 (Figures 1C and 3C).
The abovementioned results are indicative of CAV1-mediate
regulation of a variety of cellular functions, notably those that are
downstream of TCR and integrin pathways of which CAV1 serves
as the scaffold. Given these results, we were able to make predic-
tions about protein expression in relation to CAV1 and test them
in the laboratory using in vitro and in vivo experiments. For exam-
ple, we observed that CAV1 is involved in the integrin signaling
pathway that ultimately activates the mitogen-activated protein
kinase (MAPK) cascade (Figure 2); therefore, we can predict that
cellular proliferation will be decreased if CAV1 is knocked down.
VERIFICATION OF IN SILICO PREDICTIONS IN VIVO
To determine differences in morphology between CAV1−/−and
wild-type mice, we examined tissues (including lymph nodes
and spleen) stained with hematoxylin and eosin and observed
no differences were observed in tissue architecture. Lymphoid
organs (lymph nodes and spleen) were selected for their robust-
ness of CD4+ T-cells, and liver was used as a control for tis-
sue histology (i.e., to ensure mice were disease free). Further-
more, immunohistochemistry was utilized to biologically validate
in silico predictions from our CD4+ T-cell model.
Top hit proteins: GATA3, RAC1, CD26, and BCL10 were
selected for IHC to validate the model (Figure 4). RAC1 and
GATA3 showed upregulation in the lymph node tissue of the
CAV1−/−mice. We observed CD26 and BCL10 to be upregu-
lated in both the lymph node and spleen tissues of the CAV1−/−
mice. We also stained for CD28, as it is a well-characterized co-
stimulatory protein involved in T-cell activation. CD28 showed
no differential expression between the wild-type and CAV1−/−
mice.
Based on predictions from the model (Figure 3C), we investi-
gated the differential expression,hierarchal clustering (Figure 5A),
and regression analyses (Figure 5B) of top hit proteins using
microarray data from ATL cases (GSE55851). Comparison of
ATL subtypes for identification of potential molecular signa-
tures in relation to CAV1 expression reveal seven molecules,
including CAV1, clustered together based upon gene expression
profiles following hierarchal clustering (Figure 5A). We observed
a positive correlation (R = 0.78), with distinct signatures dis-
played between healthy, asymptomatic, smoldering, chronic, and
acute patients (Figure 5B). Pearson regression analyses were con-
ducted between each molecule of interest in relation to CAV1
expression across ATL subtypes (Figure 5B). Specifically, we
FIGURE 4 | Immunohistochemistry from murine model validation of
in silico-predicted differentially expressed molecules with CAV1
knockout. Molecules downstream of CAV1 that were predicted to be
affected with CAV1 knockout using the Cell Collective were validated using
lymphoid tissue histology from wild-type (WT) C57Bl/6 mice and CAV1−/−
mice.The corresponding hematoxylin and eosin preparations are included in
the top panel for morphological orientation; all tissues were sectioned
subsequently.
Frontiers in Immunology | Tumor Immunity
December 2014 | Volume 5 | Article 599 | 6
Conroy et al.
Role of CAV1 in CD4+ T-lymphocytes
FIGURE 5 | Verification and comparison of in silico results with gene
expression profiling from adultT-cell leukemia-lymphoma (ATL). (A)
Uncentered average-linkage correlation of fold-change values from top
affected proteins in ATL patients. The yellow-boxed region represents a
CAV1-associated molecular signature (R = 0.78). Healthy (n = 3),
asymptomatic (ASYM) (n = 2), smoldering (SMLD) (n = 2), chronic (CHRN)
(n = 1), and acute (ACUT) (n = 2) cases are shown. A minimum of two
samples were taken from each patient for microarray analyses. (B) Pearson
regression analyses of top affected proteins in relation to CAV1 expression
across ATL subtypes.
observed strong correlations between CAV1 and the follow-
ing molecules: BCL10 (R = 0.947), DEC2/BHLHB3 (R = 0.782),
SHP2/PTPN11 (R = 0.742), and GATA3 (R = 0.694). Conversely,
SOS1 (R = −0.981), FYN (R = −0.949), SOS2 (R = -0.825), and
CD26 (R = −0.740) were most negatively correlated to CAV1
expression. These data corroborate those described and simu-
lated in the model (Figure 3); therefore, it is translated to in vivo,
leukemic conditions.
DISCUSSION
Herein, we present a comprehensive, computational model of a
CD4+ T-cell, including CAV1 regulatory pathways. This model
incorporates experimentally validated interactions to posit the role
of CAV1 in healthy CD4+ cells and CD4+ cells in the context
of T-cell leukemia/lymphoma (i.e., when the immune response
is skewed). CD4+ T-cells are a vital component of the immune
system, as they protect against cancer, infection, and play a role
in autoimmunity. CAV1 has been shown to be upregulated in
numerous types of malignancies. Consequently, we built a CD4+
T-cell model to better understand basic T-cell biology and to
address the role(s) of CAV1 within immunocompetent versus
immunocompromised conditions. Inclusively, we investigated the
role of CAV1 in the regulation of cellular processes, including
cell cycle progression, cell proliferation, actin polymerization, and
immune synapse formation. This model was successfully con-
structed using the Cell Collective platform that allows users to
build cellular models capable of mimicking actual cellular sys-
tems in the laboratory (19–22, 38). The accuracy of the model
was then successfully validated through the comparison of simu-
lations with well-established, global input–output relationships
as previously observed experimentally. Most importantly, new
in silico predictions were validated in vitro/in vivo using both
murine models and gene expression profiles from patients with
a T-cell leukemia due to the previously observed role of Cav1 in
lymphocytes (39–46).
Using Cell Collective, we were able to perform virtual experi-
ments in order to make predictions as to how a CD4+ T-cell would
behave when the expression levels of CAV1 were altered and when
CAV1 was knocked down. These experiments provided insight as
to which proteins, and ultimately which cellular functions, might
be regulated by CAV1. Based on in silico results, we observed
that BCL10, CD26, FYN, CARMA1, SHC1, SOS, SHP2, NOS2A,
GRB2, and GATA3 are strongly influenced by CAV1 expression
(Figure 3C). Consequently, the expression of these molecules
in vivo was investigated using cluster analyses of microarray data to
measure gene expression (Figure 5). Finally,four key proteins were
selected for further verification of the predictions of the model
using immunohistochemistry on mouse tissue with and without
Cav1 (Figure 4).
In addition to the four key proteins, CD28 was chosen because
it is known to regulate T-cell activation independent of CAV1
expression (15). RAC1 and GATA3 expression were upregulated
in the lymph node tissue of the CAV1−/−mice. These results were
expected based on studies showing the role of CAV1 in lymphoid
tissue (9). We observed both CD26 and BCL10 upregulation,espe-
cially in the germinal centers of the lymph node and spleen tissue
of the CAV1−/−mice. These validations of the in silico predictions
show that the CD4+ T-cell model is biologically relevant. These
validations of the in silico predictions show that the CD4+ T-cell
model is biologically relevant. Therefore, we suggest that based on
www.frontiersin.org
December 2014 | Volume 5 | Article 599 | 7
Conroy et al.
Role of CAV1 in CD4+ T-lymphocytes
the validations in vivo the T-cell in silico model is predictive of
biological processes.
We then translated the in silico predictions to gene expression
profiles of patients with a T-cell malignancy (i.e., ATL). Of the
top 15 most differentially expressed molecules in CAV1-mediated
pathways, the top 4 most highly correlated molecules to CAV1
expression were BCL10,DEC2,SHP2,and GATA3 (R value > 0.74;
Figure 5B). Obstinately, SOS1, FYN, SOS2, and CD26 were neg-
atively correlated with CAV1 (R < 0.74; Figure 5B). Additionally,
we observed unique clustering of these 15 conserved molecules
across subtypes of ATL patients. Interestingly, there was a distin-
guishable differential expression of MALT1 in asymptomatic ATL
cases as compared with healthy individuals (Figure 5A). There-
fore, further investigation into the plausibility of these correlated
molecules being used for diagnostic purposes is warranted. We
hope that our other ongoing studies will shed light as to their
role in ATL. In short, our data regarding the role of CAV1 in cell
signaling (as demonstrated using an in silico software and subse-
quently validated experimentally) corroborate that of the existing
literature.
Specifically, CAV1 participates in processes including actin
polymerization, cell proliferation, and cell survival (9, 12, 15,
47–49).
The usefulness of the in silico approach combined with that of
in vivo/in vitro approaches provide rapid information as to the
cell signaling networks in healthy and leukemic cells. There are
currently many therapies being used to treat leukemia that tar-
get specific proteins in order to inhibit cellular pathways. These
treatment modalities are advanced when comprehensive, molecu-
lar models allow the researcher to observe the direct mechanism
of action of gene targeting as well as downstream consequences of
gene/protein knockdown. The CD4+ T-cell model will hopefully
be able to provide insight to for both T-cell biology (as demon-
strated herein) as well as possible targets for lymphocytic leukemia
treatments. The importance of in silico approaches combined with
immunoinformatics as well as in vivo validation cannot be under-
stated (18). With the often-prohibitive cost of drug design, it is
imperative to use computational approaches to derive and test
hypotheses. Current therapy regimens for T-cell malignancies can
bemodeledinsilico initiallyinanefforttounderstandmechanisms
and potential outcomes.
A comprehensive model of the CD4+ cells has the potential to
providesubstantialinsightintocancertreatment,immunotherapy,
and cellular biology. This twofold approach incorporating in silico
and in vivo investigations has the potential to translate diagnostics
and therapeutic targets from bench to bedside.
AUTHOR CONTRIBUTIONS
Brittany D. Conroy, Tyler A. Herek, and Timothy D. Shew are
equally contributing first authors. Brittany D. Conroy wrote the
paper, built the model, and did the simulations and verifications.
Timothy D. Shew and Tyler A. Herek did bioinformatics studies,
immunohistochemistry, verifications, and wrote the paper. Joshua
J. Larson assisted in bioinformatics studies and mouse work. Laura
Allen and Matthew Latner built the model, ran validations, and
performed data analyses. Paul H. Davis provided suggestions for
experimental design and helped implement experiments. Chris-
tine E. Cutucache and Tomáš Helikar conceptualized and designed
the experiment, assisted in implementation, oversaw evaluation,
and wrote the paper.
ACKNOWLEDGMENTS
We thank Andrew Pulfer for his work in helping to construct the
initial CD4+ T-cell model. Thanks to the Tissue Core Facility at
the University of Nebraska Medical Center for assistance with his-
tology. Additionally, we thank Nebraska NASA for Fellowships
(Brittany D. Conroy and Christine E. Cutucache), the University
of Nebraska at Omaha’s Sponsored Programs Office for internal
funding through an UCRCA grant (Tomáš Helikar and Christine
E. Cutucache),and thanks to Dr. George Haddix and the Nebraska
University Foundation.
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found online at
http://www.frontiersin.org/Journal/10.3389/fimmu.2014.00599/
abstract
REFERENCES
1. Mercier I, Jasmin JF, Pavlides S, Minetti C, Flomenberg N, Pestell RG, et al.
Clinical and translational implications of the caveolin gene family: lessons from
mouse models and human genetic disorders. Lab Invest (2009) 89(6):614–23.
doi:10.1038/labinvest.2009.23
2. Lajoie P, Nabi IR. Lipid rafts, caveolae, and their endocytosis. Int Rev Cell Mol
Biol (2010) 282:135–63. doi:10.1016/S1937-6448(10)82003-9
3. Kato K, Hida Y, Miyamoto M, Hashida H, Shinohara T, Itoh T, et al. Over-
expression of caveolin-1 in esophageal squamous cell carcinoma correlates
with lymph node metastasis and pathologic stage. Cancer (2002) 94(4):929–33.
doi:10.1002/cncr.10329
4. Williams T, Lisanti M. Caveolin-1 in oncogenic transformation, cancer, and
metastasis. Am J Physiol (2005) 288(3):C494–506. doi:10.1152/ajpcell.00458.
2004
5. Yeh D, Chen C, Sun MZ, Shao S, Hao L, Song Y, et al. Caveolin-1 is an impor-
tant factor for the metastasis and proliferation of human small cell lung cancer
NCI-H446 cell. Anat Rec (2009) 292(10):1584–92. doi:10.1002/ar.20974
6. Wiechen K, Sers C, Agoulnik A, Arlt K, Dietel M, Schlag PM, et al. Down-
regulation of caveolin-1, a candidate tumor suppressor gene, in sarcomas. Am J
Pathol (2001) 158(3):833–9. doi:10.1016/S0002-9440(10)64031-X
7. Capozza, F, Williams, TM, Schubert, W, McClain, S, Bouzahzah, B, Sotgia, F,
et al. Absence of caveolin-1 sensitizes mouse skin to carcinogen-induced epi-
dermal hyperplasia and tumor formation. Am J Pathol (2003) 162(6):2029–39.
doi:10.1016/S0002-9440(10)64335-0
8. Kim EJ, Sohn JH, Kim M, Chae SW, Lee HS, Coo EY, et al. The loss of expres-
sion of caveolin-1 in gastrointestinal stromal tumors. Korean J Pathol (2005)
39(5):338–44.
9. Gilling CE, Mittal AK, Chaturvedi N, Iqbal J, Aoun P, Bierman P, et al. Lymph
node-induced immune tolerance in chronic lymphocytic leukaemia: a role for
caveolin-1. Br J Haematol (2012) 158(2):216–31. doi:10.1111/j.1365-2141.2012.
09148.x
10. Gilling CE, Mittal AK, Joshi SS. Immune synapse malformation: role for CAV1
and PAK1. FASEB J (2012) 26:522.7. doi:10.1096/fj.1530-6860
11. Proust R, Bertoglio J, Gesbert F. The adaptor protein SAP directly associates
with CD3ζ chain and regulates T cell receptor signaling. PLoS One (2012) 7:8.
doi:10.1371/journal.pone.0043200
12. Ohnuma K, Uchiyama M, Yamochi T, Nishibashi K, Hosono O, Takahashi
N, et al. Caveolin-1 triggers T-cell activation via CD26 in association with
CARMA1. J Biol Chem (2007) 282(13):10117–31. doi:10.1074/jbc.M609157200
13. Kortum RL, Fernandez MR, Costanzo-Garvey DL, Johnson HJ, Fisher KW,Volle
DJ, et al. Caveolin-1 is required for kinase suppressor of Ras 1 (KSR1)-mediated
ERK1/2 activation, H-rasv12-induced senescence and transformation. Mol Cell
Biol (2014) 34(18):3461–72. doi:10.1128/MCB.01633-13
Frontiers in Immunology | Tumor Immunity
December 2014 | Volume 5 | Article 599 | 8
Conroy et al.
Role of CAV1 in CD4+ T-lymphocytes
14. Ishii T, Ohnuma K, Murakami A, Takasawa N, Kobayashi S, Dang NH, et al.
CD26-mediated signaling for T cell activation occurs in lipid rafts through its
association with CD45RO. Proc Natl Acad Sci U S A (2001) 98(21):12138–43.
doi:10.1073/pnas.211439098
15. Tomassian T,Humphries LA,Liu SD,Silva O,Brooks DG,Miceli MC. Caveolin-1
orchestrates TCR synaptic polarity, signal specificity, and function in CD8 T
cells. J Immunol (2011) 187(6):2993–3002. doi:10.4049/jimmunol.1101447
16. Burger JA, Kipps TJ. Chemokine receptors and stromal cells in the homing and
homeostasis of chronic lymphocytic leukemia B cells. Leuk Lymphoma (2002)
43(3):461–6. doi:10.1080/10428190290011921
17. Cutucache CE. Tumor-induced host immunosuppression: special focus on CLL.
Int Immunopharmacol (2013) 17(1):35–41. doi:10.1016/j.intimp.2013.05.021
18. Tomar N, De RK. Immunoinformatics: a brief review. Methods Mol Biol (2014)
1184:23–55. doi:10.1007/978-1-4939-1115-8_3
19. HelikarT,KonvalinaJ,HeidelJ,RogersJA.Emergentdecision-makinginbiologi-
cal signal transduction networks. Proc NatlAcad Sci U SA (2008) 105(6):1913–8.
doi:10.1073/pnas.0705088105
20. Helikar T, Kowal B, McClenathan S, Bruckner M, Rowley T, Madrahimov A,
et al. The cell collective: toward an open and collaborative approach to systems
biology. BMC Syst Biol (2012) 6(1):96. doi:10.1186/1752-0509-6-96
21. Madrahimov A, Helikar T, Kowal B, Lu G, Rogers J. Dynamics of influenza virus
and human host interactions during infection and replication cycle. Bull Math
Biol (2013) 75(6):988–1011. doi:10.1007/s11538-012-9777-2
22. Helikar T, Kochi N, Kowal B, Dimri M, Naramura M, Raja SM, et al. A com-
prehensive, multi-scale dynamical model of ErbB receptor signal transduction
in human mammary epithelial cells. PLoS One (2013) 8:4. doi:10.1371/journal.
pone.0061757
23. Morris MK, Saez-Rodriguez J, Sorger PK, Lauffenburger DA. Logic-based
models for the analysis of cell signaling networks. Biochemistry (2010)
49(15):3216–24. doi:10.1021/bi902202q
24. Helikar T, Kowal B, Madrahimov A, Shrestha M, Pedersen J, Limbu K, et al. Bio-
Logic Builder: a nontechnical tool for building dynamical, qualitative models.
PLoS One (2012) 7(10):e46417. doi:10.1371/journal.pone.0046417
25. Naldi A, Carneiro J, Chaouiya C, Thieffry D. Diversity and plasticity of Th cell
types predicted from regulatory network modelling. PLoS Comput Biol (2010)
6(9):e1000912. doi:10.1371/journal.pcbi.1000912
26. Chaouiya C, Bérenguier D, Keating SM, Naldi A, van Iersel MP, Rodriguez N,
et al. SBML qualitative models: a model representation format and infrastruc-
ture to foster interactions between qualitative modelling formalisms and tools.
BMC Syst Biol (2013) 7:135. doi:10.1186/1752-0509-7-135
27. Alonso MA, Millán J. The role of lipid rafts in signaling and membrane traffick-
ing in T lymphocytes. J Cell Sci (2001) 114:3957–65.
28. Kumar CC. Signaling by integrin receptors. Oncogene (1998) 17:1365–73.
doi:10.1038/sj.onc.1202172
29. Shi CS,Kehrl JR. PYK2 links G(q)alpha and G(13)alpha signaling to NF-kappa B
activation. J Biol Chem (2001) 276(34):31845–50. doi:10.1074/jbc.M101043200
30. McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, Change F,
et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transfor-
mation and drug resistance. Biochim Biophys Acta (2007) 1773(8):1263–84.
doi:10.1016/j.bbamcr.2006.10.001
31. Lundin Brockdorff J, Woetmann A, Mustelin T, Kaltoft K, Zhang Q, Wasik MA,
et al. SHP2 regulates IL-2 induced MAPK activation, but not STAT3 or STAT5
tyrosine phosphorylation, incutaneous T cell lymphoma cells. Cytokine (2002)
4:141–7. doi:10.1006/cyto.2002.1986
32. Burchill MA, Yang J, Vogtenhuber C, Blazar BR, Farrar MA. IL-2 receptor beta-
dependent STAT5 activation is required for the development of Foxp3+ regula-
tory T cells. J Immunol (2007) 178:280–90. doi:10.4049/jimmunol.178.1.280
33. Hofmann C, Shepelev M, Chernoff J. The genetics of Pak. J Cell Sci (2004)
117(19):4343–54. doi:10.1242/jcs.01392
34. IqbalJ,KucukC,DeleeuwRJ,SrivastavaG,TamW,GengH,et al.Genomicanaly-
ses reveal global functional alterations that promote tumor growth and novel
tumor suppressor genes in natural killer-cell malignancies. Leukemia (2009)
23(6):1139–51. doi:10.1038/leu.2009.3
35. Mittal AK, Gilling CE, Iqbal J, Bociek RG, Aoun P, Bierman P, et al. Clinical
heterogeneity of CLL: role for immune dysregulation mediated by the lymph
node microenvironment. Blood (2009) 546:112.
36. Helikar T, Rogers JA. ChemChains: a platform for simulation and analysis of
biochemical networks aimed to laboratory scientists. BMC Syst Biol (2009) 3:58.
doi:10.1186/1752-0509-3-58
37. Kobayashi S, Nakano K, Watanabe E, Ishigaki T, Ohno N, Yuji K, et al. CADM1
expression and stepwise downregulation of CD7 are closely associated with
clonal expansion of HTLV-1-infected cells in adult T-cell leukemia/lymphoma.
Clin Cancer Res (2014). doi:10.1158/1078-0432.CCR-13-3169
38. Todd R, Helikar T. Ergodic sets as cell phenotype of budding yeast cell cycle.
PLoS One (2012) 7(10):e45780. doi:10.1371/journal.pone.0045780
39. Fischer KD,KongYY,Nishina H,Tedford K,Marengere LEM,Kozieradzki I,et al.
Vav is a regulator of cytoskeletal reorganization mediated by the T-cell receptor.
Curr Biol (1998) 8(10):554–62. doi:10.1016/S0960-9822(98)70224-6
40. Wary KK, Mariotti A, Zurzolo C, Giancotti FG. A requirement for caveolin-1
and associated kinase Fyn in integrin signaling and anchorage-dependent cell
growth. Cell (1998) 94(5):625–34. doi:10.1016/S0092-8674(00)81604-9
41. Edwards DC, Sanders LC, Bokoch GM, Gill GN. Activation of LIM-kinase by
Pak1 couples Rac/Cdc42 GTPase signaling to actin cytoskeletal dynamics. Nat
Cell Biol (1999) 1(5):253–9. doi:10.1038/12963
42. Egawa T, Albrecht B, Favier B, Sunshine MJ, Mirchandani K, O’Brien W, et al.
Requirement for CARMA1 in antigen receptor-induced NF-κB activation and
lymphocyte proliferation. Curr Biol (2003) 13(14):1252–8. doi:10.1016/S0960-
9822(03)00491-3
43. Meffert MK, Chang JM, Wiltgen BJ, Fanselow MS, Baltimore D. NF-κB func-
tions in synaptic signaling and behavior. Nat Neurosci (2003) 6(10):1072–8.
doi:10.1038/nn1110
44. Dutt P, Nguyen N, Toksoz D. Role of Lbc RhoGEF in Ga12/13-induced signals
to Rho GTPase. Cell Signal (2004) 16(2):201–9. doi:10.1016/S0898-6568(03)
00132-3
45. Görgün G, Holderried TA, Zahrieh D, Neuberg D, Gribben JG. Chronic lym-
phocytic leukemia cells induce changes in gene expression of CD4 and CD8 T
cells. J Clin Invest (2005) 115(7):1797–805. doi:10.1172/JCI24176
46. Garcia-Closas M, Troester MA, Qi Y, Langerød A, Yeager M, Lissowska J,
et al. Common genetic variation in GATA-binding protein 3 and differential
susceptibility to breast cancer by estrogen receptor alpha tumor status. Can-
cer Epidemiol Biomarkers Prev (2007) 16(11):2269–75. doi:10.1158/1055-9965.
EPI-07-0449
47. Lee H, Volonte D, Galbiati F, Iyengar P, Lublin DM, Bregman DB, et al. Con-
stitutive and growth factor-regulated phosphorylation of caveolin-1 occurs at
the same site (Tyr-14) in vivo: identification of a c-Src/Cav-1/Grb7 signaling
cassette. Mol Endocrinol (2000) 14(11):1750–75. doi:10.1210/mend.14.11.0553
48. Sasahara Y, Rachid R, Byrne MJ, de la Fuente MA, Abraham RT, Ramesh
N, et al. Mechanism of recruitment of WASP to the immunological synapse
and of its activation following TCR ligation. Mol Cell (2002) 10(6):1269–81.
doi:10.1016/S1097-2765(02)00728-1
49. Thome M. CARMA1, BCL-10 and MALT1 in lymphocyte development and
activation. Nat Rev Immunol (2004) 4(5):348–59. doi:10.1038/nri1352
Conflict of Interest Statement: Tomáš Helikar is a founder and scientific advisor
to Discovery Collective, Inc. Discovery Collective holds a license to use the Cell
Collective software. All other authors declare no conflict of interest.
Received: 25 August 2014; accepted: 07 November 2014; published online: 05 December
2014.
Citation: Conroy BD, Herek TA, Shew TD, Latner M, Larson JJ, Allen L, Davis PH,
Helikar T and Cutucache CE (2014) Design, assessment, and in vivo evaluation of
a computational model illustrating the role of CAV1 in CD4+ T-lymphocytes. Front.
Immunol. 5:599. doi: 10.3389/fimmu.2014.00599
This article was submitted to Tumor Immunity, a section of the journal Frontiers in
Immunology.
Copyright © 2014 Conroy, Herek, Shew, Latner, Larson, Allen, Davis, Helikar and
Cutucache. This is an open-access article distributed under the terms of the Creative
Commons Attribution License (CC BY). The use, distribution or reproduction in other
forums is permitted, provided the original author(s) or licensor are credited and that
the original publication in this journal is cited, in accordance with accepted academic
practice. No use, distribution or reproduction is permitted which does not comply with
these terms.
www.frontiersin.org
December 2014 | Volume 5 | Article 599 | 9
|
25538703
|
FYN = ( CAV1_scaffold ) OR ( CD3 AND ( ( ( TCR ) ) ) )
PKA = ( cAMP )
MEK3 = ( MEKK4 )
FOXP3 = ( ( ( SMAD3 AND ( ( ( STAT5 AND NFAT ) ) ) ) AND NOT ( STAT1 ) ) AND NOT ( STAT3 AND ( ( ( RORGT ) ) ) ) ) OR ( NFAT AND ( ( ( FOXP3 AND STAT5 ) ) ) )
IL17 = ( ( ( ( NFAT AND ( ( ( proliferation AND STAT3 AND NFKB AND RORGT ) ) ) ) AND NOT ( STAT5 AND ( ( ( FOXP3 ) ) ) ) ) AND NOT ( STAT6 AND ( ( ( FOXP3 ) ) ) ) ) AND NOT ( STAT1 AND ( ( ( FOXP3 ) ) ) ) )
Lck = ( CD28 ) OR ( JAK3 AND ( ( ( IL2RB ) ) ) ) OR ( CD4 )
IL4 = ( IRF4 ) OR ( ( ( ( GATA3 AND ( ( ( proliferation AND NFAT ) ) ) ) AND NOT ( FOXP3 ) ) AND NOT ( TBET AND ( ( ( RUNX3 ) ) ) ) ) AND NOT ( IRF1 ) )
IL23 = ( NFAT AND ( ( ( proliferation AND STAT3 ) ) ) )
TGFB = ( FOXP3 AND ( ( ( proliferation AND NFAT ) ) ) )
Vav = ( SLP-76 )
IL4R_HIGH = ( IL4 AND ( ( ( IL4RA_HIGH AND CGC ) ) ) ) OR ( IL4_e AND ( ( ( IL4RA_HIGH AND CGC ) ) ) )
GSK-3b = ( NOT ( ( AKT ) ) ) OR NOT ( AKT )
was = ( Src )
IL2 = ( ( ( NFAT AND ( ( ( NOT FOXP3 ) ) ) ) AND NOT ( TBET AND ( ( ( NFKB ) ) ) ) ) AND NOT ( STAT5 AND ( ( ( STAT6 ) ) ) ) ) OR ( ( ( NFKB ) AND NOT ( TBET AND ( ( ( NFKB ) ) ) ) ) AND NOT ( STAT5 AND ( ( ( STAT6 ) ) ) ) )
C3G = ( Crk )
IL9R = ( IL9_e ) OR ( JAK3 )
Nck = ( SLP-76 )
FAK_Tyr397 = ( Bintegrin )
PDK1 = ( PIP3_345 )
MKK7 = ( TAK1 )
STAT3 = ( IL21R ) OR ( IL27R ) OR ( IL6R ) OR ( IL23R ) OR ( IL10R )
SMAD3 = ( TGFBR )
Profilin = ( RIAM )
Calcineurin = ( Ca2+ )
IFNBR = ( IFNB_e )
IL2RA = ( FOXP3 AND ( ( ( NFAT ) ) ) ) OR ( STAT5 AND ( ( ( NFAT ) ) ) ) OR ( SMAD3 AND ( ( ( NFAT ) ) ) ) OR ( NFKB AND ( ( ( NFAT ) ) ) )
HLX = ( TBET )
Shc1 = ( FYN ) OR ( IL2RB AND ( ( ( IL2R ) ) ) )
Cdc42 = ( C3G ) OR ( RhoGEF )
IL4RA = ( NOT ( ( STAT5_HIGH ) ) ) OR NOT ( STAT5_HIGH )
Tyk2 = ( IL12RB1 AND ( ( ( IL12RB2 ) ) ) )
IL2R_HIGH = ( IL2 AND ( ( ( IL2RB AND CGC AND IL2RA ) ) ) ) OR ( IL2_e AND ( ( ( IL2RB AND CGC AND IL2RA ) ) ) )
N_WASP = ( Cdc42 ) OR ( Nck AND ( ( ( Vav ) ) ) )
IL22 = ( STAT4 ) OR ( STAT5 ) OR ( STAT1 ) OR ( STAT3 )
RhoA = ( CAV1_scaffold ) OR ( RhoGEF )
PKC = ( DAG )
JAK3 = ( IL2R )
CD28 = ( APC ) OR ( B7 )
IL4RA_HIGH = ( STAT5_HIGH )
NFKB = ( NOT ( ( FOXP3 ) OR ( IKB ) ) ) OR NOT ( IKB OR FOXP3 )
IL12RB2 = ( IL12_e )
F_Actin = ( Arp2_3 AND ( ( ( G_Actin ) ) ) )
ROCK = ( RhoA )
MEKK4 = ( GADD45B AND ( ( ( GADD45G ) ) ) )
CARMA1 = ( CD26 ) OR ( PKC )
ERM = ( STAT4 )
Galpha_Q = ( Galpha_QL )
IL10 = ( NFAT AND ( ( ( GATA3 OR STAT3 ) AND ( ( ( proliferation ) ) ) ) ) )
ATF2 = ( P38 )
IKB = ( NOT ( ( IKKcomplex ) ) ) OR NOT ( IKKcomplex )
MEK1_2 = ( RAF1 ) OR ( BRAF ) OR ( PAK )
GalphaS_R = ( GalphaS_L )
GATA3 = ( ( STAT6 ) AND NOT ( TBET ) ) OR ( Dec2 )
rac1 = ( was ) OR ( Crk AND ( ( ( Paxillin ) ) ) ) OR ( NOS2A ) OR ( Vav )
CD4 = ( TCR AND ( ( ( MHC_II AND CD3 ) ) ) )
Src = ( Bintegrin ) OR ( FAK_Tyr397 )
IL23R = ( IL23 AND ( ( ( GP130 AND STAT3 AND IL12RB1 AND RORGT ) ) ) ) OR ( IL23_e AND ( ( ( GP130 AND STAT3 AND IL12RB1 AND RORGT ) ) ) )
NOS2A = ( CAV1_scaffold )
TRAF6 = ( IRAK1 )
GFI1 = ( TCR ) OR ( STAT6 )
Dec2 = ( GATA3 )
Cas = ( FAK_576_577 AND ( ( ( Bintegrin ) ) ) )
Arp2_3 = ( WAVE-2 ) OR ( N_WASP )
SLP-76 = ( ZAP-70 ) OR ( Gads )
Gads = ( LAT )
IFNGR = ( IFNG_e AND ( ( ( IFNGR2 AND IFNGR1 ) ) ) ) OR ( IFNG AND ( ( ( IFNGR2 AND IFNGR1 ) ) ) )
Grb2 = ( Shc1 ) OR ( LAT )
IL4R = ( IL4 AND ( ( ( IL4RA AND CGC ) ) ) ) OR ( IL4_e AND ( ( ( IL4RA AND CGC ) ) ) )
PI3K = ( CD28 AND ( ( ( ICOS ) ) ) ) OR ( SHP2 ) OR ( IL2R ) OR ( GAB2 ) OR ( Ras ) OR ( FAK_576_577 )
LIMK = ( PAK ) OR ( ROCK )
SOCS1 = ( STAT6 ) OR ( STAT3 )
ERK = ( MEK1_2 )
SOCS3 = ( STAT3 )
RASgrp = ( DAG )
NFAT = ( CD28 AND ( ( ( TCR ) ) ) ) OR ( ( Calcineurin AND ( ( ( P38 ) ) ) ) AND NOT ( GSK-3b ) ) OR ( TCR AND ( ( ( CD28 ) ) ) )
IL21R = ( IL21 AND ( ( ( GP130 AND CGC ) ) ) ) OR ( IL21_e AND ( ( ( GP130 AND CGC ) ) ) )
IL6R = ( GP130 AND ( ( ( IL6_e AND IL6RA ) ) ) )
AKT = ( PDK1 )
RAF1 = ( Ras )
Paxillin = ( FAK_576_577 )
LAT = ( ZAP-70 )
ITAMS = ( Lck )
PIP3_345 = ( PI3K )
Sos = ( Grb2 )
CAV1_scaffold = ( Src ) OR ( CAV1_ACTIVATOR ) OR ( Bintegrin )
MEK4 = ( MEKK4 )
BCL10_Malt1 = ( CARMA1 )
STAT4 = ( ( JAK2 ) AND NOT ( GATA3 ) ) OR ( ( P38 AND ( ( ( Tyk2 ) ) ) ) AND NOT ( GATA3 ) )
G_Actin = ( Profilin )
JNK = ( rac1 AND ( ( ( Crk ) ) ) ) OR ( MEK4 ) OR ( MKK7 )
CD26 = ( CAV1_scaffold )
ITK = ( SLP-76 )
IRF4 = ( GATA3 )
adenyl_cyclase = ( GalphaS_R )
GAB2 = ( Shc1 AND ( ( ( Grb2 ) ) ) )
proliferation = ( proliferation ) OR ( STAT5_HIGH )
IRSp53 = ( rac1 )
IL21 = ( NFAT AND ( ( ( proliferation AND STAT3 ) ) ) )
PAK = ( rac1 ) OR ( Cdc42 ) OR ( Nck )
MEK6 = ( MEKK4 )
FAK_576_577 = ( FAK_Tyr397 AND ( ( ( Src ) ) ) )
Galpha_iR = ( Galpha_iL )
IL12RB1 = ( IRF1 ) OR ( IL12_e )
GADD45G = ( IL12_e ) OR ( CD3 )
IFNG = ( ( ( STAT4 AND ( ( ( proliferation AND NFAT ) ) ) ) AND NOT ( FOXP3 ) ) AND NOT ( STAT3 ) ) OR ( ( ( ATF2 ) AND NOT ( FOXP3 ) ) AND NOT ( STAT3 ) ) OR ( ( ( AP1 AND ( ( ( STAT4 ) ) ) ) AND NOT ( FOXP3 ) ) AND NOT ( STAT3 ) ) OR ( ( ( RUNX3 AND ( ( ( proliferation AND TBET AND NFAT ) ) ) ) AND NOT ( FOXP3 ) ) AND NOT ( STAT3 ) ) OR ( ( ( HLX ) AND NOT ( FOXP3 ) ) AND NOT ( STAT3 ) )
IL15R = ( CGC AND ( ( ( IL2RB AND IL15RA AND IL15_e ) ) ) )
TBET = ( ( TBET ) AND NOT ( GATA3 ) ) OR ( ( STAT1 ) AND NOT ( GATA3 ) )
EPAC = ( cAMP )
RhoGEF = ( Galpha12_13R ) OR ( FAK_576_577 )
IL2R = ( IL2 AND ( ( ( IL2RB AND CGC ) AND ( ( ( NOT IL2RA ) ) ) ) ) ) OR ( IL2_e AND ( ( ( IL2RB AND CGC ) AND ( ( ( NOT IL2RA ) ) ) ) ) )
STAT6 = ( IL4R )
STAT5_HIGH = ( IL4R_HIGH ) OR ( IL2R_HIGH )
TCR = ( APC AND ( ( ( CD28 ) ) ) )
STAT1 = ( ( IFNBR ) AND NOT ( SOCS1 ) ) OR ( ( IFNGR ) AND NOT ( SOCS1 ) ) OR ( ( IL27R ) AND NOT ( SOCS1 ) )
DAG = ( PLCb ) OR ( PLCg )
JAK1 = ( ( IL2R ) AND NOT ( SOCS3 ) ) OR ( ( JAK3 ) AND NOT ( SOCS3 ) ) OR ( ( IL9R ) AND NOT ( SOCS3 ) ) OR ( ( IL22R ) AND NOT ( SOCS3 ) )
IL10R = ( IL10 AND ( ( ( IL10RB AND IL10RA ) ) ) ) OR ( IL10_e AND ( ( ( IL10RB AND IL10RA ) ) ) )
Ras = ( Sos ) OR ( RASgrp )
IP3 = ( PLCg )
BRAF = ( Rap1 )
RIAM = ( Rap1 )
Ca2+ = ( IP3 )
IRAK1 = ( IL18R1 )
Cofilin = ( NOT ( ( LIMK ) ) ) OR NOT ( LIMK )
IL18R1 = ( IL18_e )
ZAP-70 = ( ITAMS AND ( ( ( CD3 ) ) ) )
Rap1 = ( EPAC ) OR ( C3G AND ( ( ( Crk ) ) ) ) OR ( PKA )
IRF1 = ( STAT1 )
AP1 = ( STAT4 ) OR ( JNK ) OR ( ERK )
Bintegrin = ( ECM ) OR ( TCR )
PLCb = ( Galpha_Q )
MLC = ( ROCK )
cAMP = ( adenyl_cyclase )
SYK = ( IL2R )
ICOS = ( APC )
RUNX3 = ( ( TBET ) AND NOT ( GATA3 ) )
RORGT = ( RORGT AND ( ( ( STAT3 OR TGFBR ) ) ) ) OR ( TGFBR AND ( ( ( STAT3 ) ) ) )
Galpha12_13R = ( alpha_13L )
PLCg = ( ZAP-70 ) OR ( ITK ) OR ( LAT )
NIK = ( TRAF6 )
CD3 = ( TCR )
WAVE-2 = ( IRSp53 AND ( ( ( rac1 ) ) ) )
Bcl10_Carma1_MALTI = ( BCL10_Malt1 AND ( ( ( CARMA1 ) ) ) )
GADD45B = ( IL12_e AND ( ( ( TCR ) ) ) )
IL27R = ( GP130 AND ( ( ( IL27_e AND IL27RA ) ) ) )
JAK2 = ( IL12RB1 AND ( ( ( IL12RB2 ) ) ) )
IL22R = ( IL22_e )
Crk = ( Cas ) OR ( Paxillin )
P38 = ( MEK3 ) OR ( MEK6 )
IKKcomplex = ( Bcl10_Carma1_MALTI ) OR ( NIK ) OR ( TCR )
SHP2 = ( GAB2 ) OR ( IL2RB )
STAT5 = ( IL2R ) OR ( IL4R ) OR ( SYK ) OR ( IL15R ) OR ( Lck ) OR ( JAK1 )
TAK1 = ( TRAF6 )
TGFBR = ( TGFB ) OR ( TGFB_e )
|
EDUCATION
Integrating Interactive Computational
Modeling in Biology Curricula
Tomáš Helikar1*, Christine E. Cutucache2, Lauren M. Dahlquist2, Tyler A. Herek2, Joshua
J. Larson2, Jim A. Rogers3
1 Department of Biochemistry, University of Nebraska–Lincoln, Lincoln, Nebraska, United States of America,
2 Department of Biology, University of Nebraska–Omaha, Omaha, Nebraska, United States of America,
3 Department of Mathematics, University of Nebraska–Omaha, Omaha, Nebraska, United States of America
* thelikar2@unl.edu
Abstract
While the use of computer tools to simulate complex processes such as computer circuits is
normal practice in fields like engineering, the majority of life sciences/biological sciences
courses continue to rely on the traditional textbook and memorization approach. To address
this issue, we explored the use of the Cell Collective platform as a novel, interactive, and
evolving pedagogical tool to foster student engagement, creativity, and higher-level think-
ing. Cell Collective is a Web-based platform used to create and simulate dynamical models
of various biological processes. Students can create models of cells, diseases, or pathways
themselves or explore existing models. This technology was implemented in both under-
graduate and graduate courses as a pilot study to determine the feasibility of such software
at the university level. First, a new (In Silico Biology) class was developed to enable stu-
dents to learn biology by “building and breaking it” via computer models and their simula-
tions. This class and technology also provide a non-intimidating way to incorporate
mathematical and computational concepts into a class with students who have a limited
mathematical background. Second, we used the technology to mediate the use of simula-
tions and modeling modules as a learning tool for traditional biological concepts, such as
T cell differentiation or cell cycle regulation, in existing biology courses. Results of this pilot
application suggest that there is promise in the use of computational modeling and software
tools such as Cell Collective to provide new teaching methods in biology and contribute to
the implementation of the “Vision and Change” call to action in undergraduate biology edu-
cation by providing a hands-on approach to biology.
Introduction
The enormous complexity that recent research has revealed in biological and biochemical sys-
tems has resulted in the emergence of mathematical modeling and computer simulations as an
integral part of biomedical research. This provides researchers with new tools to understand
the role of emergent properties in healthy and diseased cells, to generate new hypotheses, and
even screen potential pharmaceuticals for cross-reactivity and potential targets [1–3].
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004131
March 19, 2015
1 / 9
a11111
OPEN ACCESS
Citation: Helikar T, Cutucache CE, Dahlquist LM,
Herek TA, Larson JJ, Rogers JA (2015) Integrating
Interactive Computational Modeling in Biology
Curricula. PLoS Comput Biol 11(3): e1004131.
doi:10.1371/journal.pcbi.1004131
Editor: Joanne A. Fox, University of British
Columbia, CANADA
Published: March 19, 2015
Copyright: © 2015 Helikar et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Funding: Office of Sponsored Programs at the
University of Nebraska at Omaha via University
Committee on Research and Creative Activity grants
to TH, CC, TAH, and LD. National Institutes of Health
(#5R01DA030962) to JAR. University of Nebraska–
Lincoln to TH. The funders had no role in the
preparation of the manuscript.
Competing Interests: Tomáš Helikar and Jim A
Rogers are or have served as scientific advisors and/
or consultants to Discovery Collective.
Given the fact that the field is undergoing a shift in the basic way the functions of these dy-
namical systems/networks are understood, it is essential for biology education to evolve in
order to reflect these changes [4,5]. It is vital for students to learn about these structures and
the resultant emergent properties that are not obvious from looking at static pictures in text-
books. Furthermore, the National Science Foundation and the American Association for the
Advancement of Science have initiated a call to action, “Vision and Change” [6], that aims to
transform undergraduate biology education by incorporating computational methods and by
introducing key core competencies including simulation and modeling. A number of efforts
have already been initiated in this direction, including problem-based learning in the under-
graduate setting [7], translational approaches (i.e., having students serve as researchers in the
classrooms to investigate biological problems and identify solutions), as well as those led by
Carl Wieman of the Carl Wieman Institute [8] and other leaders in foundational learning (e.g.,
[9,10]).
Our group has also attempted to address this issue using our recently developed and re-
leased modeling platform called Cell Collective [11,12]. The platform enables scientists to cre-
ate, simulate, and analyze large-scale computational models of various biological systems
without the need to enter/modify any mathematical expressions and/or computer code. Be-
cause accessibility to modeling for a wide audience is the key ingredient of the technology, the
platform lends itself to application in a classroom setting. Specifically, students can create, sim-
ulate, and analyze then break and re-create and re-analyze dynamical models to understand
major biological processes. The collaborative nature of the Web-based environment enables
students to easily collaborate inside and outside of the classroom in a meaningful way. The
types of biological processes that can be explored with Cell Collective are virtually unlimited;
students can model biological processes including, but not limited to, cellular development, cel-
lular differentiation, cell-to-cell interactions, disease pathogenesis, the effects of various treat-
ments on disease, etc.
Herein, we discuss two different applications of the Cell Collective’s interactive technology
as a tool to facilitate hands-on, creative learning in the classroom and allow students to apply
their knowledge in real-time. The first is using Cell Collective in a dedicated course (In Silico
Biology) designed around the use of the technology, and the second involves introducing the
technology as a supplement to existing, traditional biology courses. Both applications have
been subjected to initial testing in a variety of undergraduate settings, and the results indicate
that both methods were successful in increasing both understanding of and enthusiasm for
complex biological systems in undergraduate student populations.
A New Course Designed for Integrated Learning of Biological and
Computational Concepts
In Silico Biology is a course that was designed de novo to use Cell Collective as the central tool
for teaching students complex biochemical systems by recreating them in silico. The individual
objectives of the course include helping students to expand their analytical skills and become
interested in computational sciences, learn to actively read primary journal articles, critically
analyze and interpret data, and use interactive computational models to learn about biological
networks. This is facilitated throughout the course via three major topics and strategies incor-
porated into the course:
1. Introduction of biological concepts from a systems perspective
The focus of the biological component of the In Silico Biology course is on complex networks
found in biological systems. A series of lectures at the beginning of the semester provide
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004131
March 19, 2015
2 / 9
students with the foundation of molecular biology of the cell, including the principles of intra-
and intercellular signal transduction. During this session, students also learn to think about
biochemical protein regulatory mechanisms from a holistic perspective; that is, rather than fo-
cusing on individual protein–protein interactions, students are expected to research and under-
stand the overall regulatory mechanism of a given protein while taking into account most
known interaction partners.
For example, students are required to go beyond the traditional representation of the regula-
tion of the Raf protein (Fig. 1). Raf is a key component of the mitogen activated protein kinase
(MAPK) pathway, which regulates numerous cellular functions (e.g., growth, apoptosis, etc.).
Students learn that Ras is only one of many components required to successfully activate, as
well as deactivate, Raf via a combination of biochemical events (Fig. 1B) [13,14]. In the final
part of this session, students are expected to research (from published literature) and describe
the complete regulatory mechanism of an enzyme of their choice, as a system of multiple inter-
action components. In this session, students are also introduced to Cell Collective, and are ex-
pected to create and simulate a simple pathway model such as the one illustrated in Fig. 1A, as
well as to model the regulatory dynamics of the researched enzyme. Importantly, with this ap-
proach, the students learn how to read and critically analyze primary journal articles.
(Fig. 1 adapted from [14])
2. Introduction to the dynamics of biological systems via computational
modeling
In this part of the course, students learn the principles of the technology and modeling frame-
work on which Cell Collective is based (Section 1 in S1 Text) [15–20]. This includes the differ-
ent types of representation of Boolean functions, as well as concepts of state transition graphs,
feedback loops, attractors, attractor stability, etc. All of these concepts are tied to and demon-
strated in the Cell Collective platform and applied to biological examples. Students also learn
about nonlinear dynamics such as bistability and oscillations associated with positive and nega-
tive feedback loops, respectively. By the end of this session, students are able to represent com-
plex biological regulatory mechanisms as Boolean functions and create and simulate the
dynamics of their corresponding models (by hand, as well as in Cell Collective). An example of
a biological system well-suited to the approach is bacterial chemotaxis (Section 2 in S1 Text).
3. Blurring the line between education and research: incorporating
meaningful undergraduate research experiences into the classroom
A large part of the course is devoted to a hands-on project during which students learn about a
biological system by integrating the biological and computational concepts they learned in the
course. Specifically, students select a biological network process of interest that they research,
construct a computational model representation of, and study the dynamics of the process by
simulating the model in Cell Collective.
From our experience, it is the learning-by-building approach that enables students to learn
and appreciate the diversity and complexities of biological systems. This method fosters curios-
ity from the students, which keeps them motivated and active in the project. Reading the litera-
ture with an objective to create a functional computer model forces the student to truly parse
and analyze the information contained in published papers in order to distill the underlying
logic of the system. Section 3 in S1 Text provides an example of how students learn by reading
the literature and performing virtual research.
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004131
March 19, 2015
3 / 9
Incorporating Computational Modeling in Existing Life Sciences
Courses
As it is not always possible or practical to create a complete course de novo, the Cell Collective
platform has also been used to aid in various existing undergraduate and graduate courses.
These include undergraduate/graduate (online) cancer biology, undergraduate microbiology,
and graduate (online) immunology courses.
In order to facilitate the introduction of modeling into existing courses, a series of modeling
modules were created; these modules are currently available in a new problem-based workbook
[21] focused on cancer biology. Students utilized the models that comprise the various modules
to simulate and analyze the dynamics of the biological processes as a way to visualize and rein-
force the content discussed during regular lectures. The interactive nature of the technology en-
ables students to alter any component or pathway of the process and, via instant feedback,
observe the effects of the change made to the system.
The modules are complementary to the traditional method of teaching as an interactive, dy-
namic process with learning objectives that match the covered topic. For example, from the ex-
ercises used in the cancer biology course, learning objectives focused on 1) determining the
Fig 1. Comparison of linear and systems representation of Raf regulation. A) Traditional (linear) representation of MAPK signaling [13]. B) Detailed
regulatory mechanism of Raf regulation that takes into account the role of most Raf interaction partners.
doi:10.1371/journal.pcbi.1004131.g001
Table 1. List of developed modeling modules.
Example Biological Concept Taught
Course Type/Topic
Malaria lifecycle
Microbiology
Positive feedback loops
Cancer biology
Negative feedback loops
Cancer biology
Cell cycle regulation
Cancer biology
DNA damage
Cancer biology
CD4+ T cell differentiation
Cancer biology, immunology
Cell communication networks
Cancer biology
doi:10.1371/journal.pcbi.1004131.t001
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004131
March 19, 2015
4 / 9
dynamic, complex signaling processes that regulate tumor development and tumor regression
and 2) the ability to illustrate feedback loops that contribute to tumor progression and regres-
sion after use of the Cell Collective. Learning objectives of the computational module used in
the microbiology course centered on the life cycle of Plasmodium spp., which leads to the de-
velopment of malaria. Specifically, after dynamically modeling and manipulating developmen-
tal processes of the Plasmodium lifecycle, students should be able to 1) draw and describe the
complex life cycle and 2) define “vector,” “reservoir,” and “transmission.”
Table 1 provides a list of modeling modules developed so far. As an example, one of these
modeling modules used for more effective learning is discussed next.
T cell differentiation and response to pathogen
T cell differentiation is an important concept taught in many immunology courses. Precise reg-
ulation of the differentiation process of naive CD4+ T cells (a subset of T-lymphocytes) to one
of the helper T cells or regulatory T cells (Tregs) is critical for the proper functioning of the im-
mune system. At the intracellular level, the differentiation process is regulated via a wide varie-
ty of types of signaling receptors and pathways that are mutually cross-linked and form highly
interconnected biochemical networks. Additionally, cytokines produced by each cell further
modulate the activation and behavior of neighboring cells, as well as the entire immune system
[22,23]. Hence, the complex network structures and nonlinear dynamics governing this pro-
cess, via both intra- and intercellular paths, make T cell differentiation a great candidate for an
interactive modeling approach. As such, a modeling module that mimics concepts and rela-
tionships (Fig. 2) was created and used to aid the learning of T cell differentiation in Cell
Collective.
An advantage to the availability of a tool such as Cell Collective is that students can alter ex-
ternal and internal conditions of the cell and observe real-time “output” or consequences at the
molecular and/or cellular level. For example, students are asked to simulate the model by first
activating antigen presenting cells (APCs) and naive T cells by introducing a “pathogen.” Path-
ogens can be introduced by changing a simple activity slider on the user interface (Fig. 3A). As
illustrated in Fig. 3B (left), the dynamical response to the change of the environment is imme-
diate. Students can subsequently simulate Th2 differentiation by introducing IL4 (Fig. 3B mid-
dle), as well as the effects of regulatory T cells (Treg) by activating TGF beta (Fig. 3B right). In
addition to the time-series, real-time simulation output, students can view the dynamics of the
entire model in a network representation in which each component of the model interactively
assumes different colors based on the activity level of the component (Fig. 3C).
Students are assigned a number of similar exercises to better understand the dynamics gov-
erning T cell regulation during the activation of the immune system, including positive feed-
back loops and associated bistable behaviors. Note that this model is one of many possible
computational model representations of T cell differentiation. Other logical models that in-
clude greater detail as to specific molecular interactions have been previously published by oth-
ers [24–26], and some of these are also available in Cell Collective for simulations.
Outcomes and Discussion
A number of efforts to incorporate computation into life science courses have been established.
For example, BioQuest consortium (http://www.bioquest.org) provides access to software
tools, datasets, and other materials developed by educators and developers engaged in educa-
tion and research in science. Another example includes NetLogo, a programming environment
for agent-based modeling that has been used to study dynamics of complex systems, as well as
for teaching of complex systems in many settings (middle schools, high schools, and
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004131
March 19, 2015
5 / 9
universities) [27]. Our approach provides a novel take on the implementation of problem-
based methods in life sciences in that it offers a Web-based, systems- and network-focused,
interactive, and real-time simulation-driven environment without the need for computer pro-
gramming or manipulation of complex mathematical equations.
We have used a Cell Collective “learning by modeling” approach both as a stand-alone class
and as a supplement to complement existing classes. In both cases, student outcomes were
highly positive (Section 5 in S1 Text). Future studies will include a comprehensive study using
both quasi-experimental and randomized control groups to determine the effect that use of
Cell Collective has on student understanding, long-term retention, critical thinking, applica-
tion, and overall mastery of material.
In addition to directly addressing the challenging problem of teaching students about com-
plex, highly connected networks, there is an additional benefit; it provides these students with
an opportunity to become interested in additional training in computational methods, some-
thing that is critical for the current and the next generation of biomedical researchers. Making
the class accessible for students with a wide range of skills such as biology, computer science,
mathematics, etc., creates an ample environment for learning from one another, resulting in
cross-pollination across disciplines.
Fig 2. CD4+ T Cell differentiation as modeled for classroom use.
doi:10.1371/journal.pcbi.1004131.g002
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004131
March 19, 2015
6 / 9
Furthermore, one of the major components of utilization of Cell Collective was blurring the
line between learning and research. This is a non-trivial aspect of this teaching method and, in-
deed, it is in some ways the most exciting—for both the students and potentially the instruc-
tors. In the course of their learning, students have the opportunity to be constructing the very
first model of the system they are studying or, if working on an existing model, they have the
opportunity to add information from recent literature to significantly update an existing
model. This means that students, while learning, are engaging in real research. In our applica-
tion of this teaching method, we have had a number of student-created and/or student-
initiated modeling projects that led to research findings, some of which were subsequently
presented by the students at an external research conference [28], and even accepted for publi-
cation in a peer-reviewed journal [29].
A positive consequence of this is that it is possible for faculty to further their own research
during the course of teaching the material. We have had several experiences of students who
had no knowledge of what they might be interested in studying being assigned a project in the
class that aligned with a research interest of our group. In several cases, the results were ulti-
mately useful to the group, and in subsequent semesters new students were assigned to either
re-create, significantly update, or provide fresh analysis of the model.
All students that made significant contributions to the models were included as authors on
all publications using that information. This result is a true win-win situation; faculty responsi-
ble for teaching a course have the possibility of actually furthering their research, while stu-
dents have the possibility to perform and be recognized for research participation. Real
Fig 3. Interactive simulation of a T cell differentiation model. A) Simple sliders can be used to change the activity levels of various stimuli. B) Example of
an interactive, real-time simulation. Left: Activation of Pathogen results in the stimulation of Antigen Presenting Cells and Naive T Cells. Middle: Stimulation
with IL4 results in the activation of Th2 cells. Right: Addition of TGF beta stimulates Tregs, resulting in the suppression of Th2 cells. C) Network view of the
changing dynamics during a real-time simulation. (Color range from bright red [inactive components] to bright green, which denotes full activity.)
doi:10.1371/journal.pcbi.1004131.g003
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004131
March 19, 2015
7 / 9
undergraduate research is not only a major goal for many universities, it is also very important
for any undergraduate looking for entry into graduate programs.
Supporting Information
S1 Text. Integrating interactive computational modeling in biology curricula.
(PDF)
Acknowledgments
We would like to thank Denis Thieffry for his feedback on the manuscript. We also thank all of
the student participants for providing feedback to help make the novel teaching approach and
software an exciting, high-quality learning tool.
References
1.
Kitano H (2002) Computational systems biology. Nature 420: 206–210. PMID: 12432404
2.
Arkin AP, Schaffer DV (2011) Network News: Innovations in 21st Century Systems Biology. Cell 144:
844–849. doi: 10.1016/j.cell.2011.03.008 PMID: 21414475
3.
Janes KA, Lauffenburger DA (2013) Models of signalling networks—what cell biologists can gain from
them and give to them. J Cell Sci 126: 1913–1921. doi: 10.1242/jcs.112045 PMID: 23720376
4.
Wilensky U, Reisman K (2005) Thinking Like a Wolf, a Sheep, or a Firefly: Learning Biology through
Constructing and Testing Computational Theories—An Embodied Modeling Approach. Cogn Instr 24:
171–209.
5.
Feser J, Vasaly H, Herrera J (2013) On the edge of mathematics and biology integration: improving
quantitative skills in undergraduate biology education. CBE Life Sci Educ 12: 124–128. doi: 10.1187/
cbe.13-03-0057 PMID: 23737616
6.
American Association for the Advancement of Science (AAAS) (2011) Vision and Change in Under-
graduate Biology Education. http://visionandchange.org/. Accessed 20 December 2013.
7.
Hmelo-Silver CE (2004) Problem-Based Learning: What and How Do Students Learn? Educ Psychol
Rev 16: 235–266. doi: 10.1023/B:EDPR.0000034022.16470.f3
8.
Wieman C, Deslauriers L, Gilley B (2013) Use of research-based instructional strategies: How to avoid
faculty quitting. Phys Rev Spec Top—Phys Educ Res 9: 023102. doi: 10.1103/PhysRevSTPER.9.
023102 PMID: 21599073
9.
Laight DW (2006) Attitudes to concept maps as a teaching/learning activity in undergraduate health
professional education: influence of preferred approach to learning. Med Teach 28: e64–7. doi: 10.
1080/01421590600617574 PMID: 16707287
10.
Musante S (2013) PULSE: Implementing Change within and among Life Science Departments. Biosci-
ence 63: 254–254. doi: 10.1525/bio.2013.63.4.4
11.
Helikar T, Kowal B, McClenathan S, Bruckner M, Rowley T, et al. (2012) The Cell Collective: Toward an
open and collaborative approach to systems biology. BMC Syst Biol 6: 96. doi: 10.1186/1752-0509-6-
96 PMID: 22871178
12.
Helikar T, Kowal B, Rogers JA (2013) A cell simulator platform: the cell collective. Clin Pharmacol Ther
93: 393–395. doi: 10.1038/clpt.2013.41 PMID: 23549147
13.
Krasinskas AM (2011) EGFR Signaling in Colorectal Carcinoma. Patholog Res Int 2011: 932932. doi:
10.4061/2011/932932 PMID: 21403829
14.
Dhillon AS, Kolch W (2002) Untying the regulation of the Raf-1 kinase. Arch Biochem Biophys 404:
3–9. doi: 10.1016/S0003-9861(02)00244-8 PMID: 12127063
15.
Kauffman S (1969) Homeostasis and differentiation in random genetic control networks. Nature 224:
177–178. PMID: 5343519
16.
Thomas R (1973) Boolean formalisation of genetic control circuits. J Theor Biol 42: 565–583.
17.
Chaouiya C, Berenguier D, Keating SM, Naldi A, van Iersel MP, et al. (2013) SBML qualitative models:
a model representation format and infrastructure to foster interactions between qualitative modelling
formalisms and tools. BMC Syst Biol 7: 135. doi: 10.1186/1752-0509-7-135 PMID: 24321545
18.
Helikar T, Kochi N, Konvalina J, Rogers JA (2011) Boolean Modeling of Biochemical Networks. Open
Bioinforma J 4: 16–25. doi: 10.1186/1687-4153-2011-797251 PMID: 21910924
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004131
March 19, 2015
8 / 9
19.
Naldi A, Berenguier D, Fauré A, Lopez F, Thieffry D, et al. (2009) Logical modelling of regulatory net-
works with GINsim 2.3. Biosystems 97: 134–139. doi: 10.1016/j.biosystems.2009.04.008 PMID:
19426782
20.
Saadatpour A, Albert I, Albert R (2010) Attractor analysis of asynchronous Boolean models of signal
transduction networks. J Theor Biol 266: 641–656. doi: 10.1016/j.jtbi.2010.07.022 PMID: 20659480
21.
Cutucache CE, Helikar T (2014) Cancer Biology: An inquiry based approach. Dubuque: Kendall/Hunt.
22.
Zhu J, Paul WE (2008) CD4 T cells: fates, functions, and faults. Blood 112: 1557–1569. doi: 10.1182/
blood-2008-05-078154 PMID: 18725574
23.
Murphy K (2012) Janeway’s Immunobiology. 8th ed. London and New York: Garland Science (Taylor
& Francis Group).
24.
Martínez-Sosa P, Mendoza L (2013) The regulatory network that controls the differentiation of T lym-
phocytes. Biosystems 113: 96–103. doi: 10.1016/j.biosystems.2013.05.007 PMID: 23743337
25.
Naldi A, Carneiro J, Chaouiya C, Thieffry D (2010) Diversity and plasticity of Th cell types predicted
from regulatory network modelling. PLoS Comput Biol 6: e1000912. doi: 10.1371/journal.pcbi.1000912
PMID: 20824124
26.
Mendoza L (2006) A network model for the control of the differentiation process in Th cells. Biosystems
84: 101–114. doi: 10.1016/j.biosystems.2005.10.004 PMID: 16386358
27.
Wilensky U (1999) NetLogo.
28.
Becker KM, Helikar T, Rogers JA, Grisham WE (2013) Modeling correspondence between auditory
and motor pathways in the zebra finch song system with boolean logic. Society for Neuroscience. San
Diego.
29.
Madrahimov A, Helikar T, Kowal B, Lu G, Rogers J (2013) Dynamics of Influenza Virus and Human
Host Interactions During Infection and Replication Cycle. Bull Math Biol 75(6):988–1011. doi: 10.1007/
s11538-012-9777-2 PMID: 23081726
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004131
March 19, 2015
9 / 9
|
25790483
|
lac_mRNA = ( lac_operon )
lactose_breakdown = ( ( lac_enzymes ) AND NOT ( betaGal_LOF_mutation ) )
lac_repressor = NOT ( ( allolactose ) )
allolactose = ( lactose )
CAP = ( cAMP )
lac_operon = ( ( CAP ) AND NOT ( lac_repressor ) )
cAMP = NOT ( ( glucose ) OR ( lactose_breakdown ) )
lac_enzymes = ( lac_mRNA )
|
Iron acquisition and oxidative stress response
in aspergillus fumigatus
Brandon et al.
Brandon et al. BMC Systems Biology (2015) 9:19
DOI 10.1186/s12918-015-0163-1
Brandon et al. BMC Systems Biology (2015) 9:19
DOI 10.1186/s12918-015-0163-1
RESEARCH ARTICLE
Open Access
Iron acquisition and oxidative stress response
in aspergillus fumigatus
Madison Brandon1,2*, Brad Howard3,4, Christopher Lawrence3,4 and Reinhard Laubenbacher2,5,6
Abstract
Background: Aspergillus fumigatus is a ubiquitous airborne fungal pathogen that presents a life-threatening health
risk to individuals with weakened immune systems. A. fumigatus pathogenicity depends on its ability to acquire iron
from the host and to resist host-generated oxidative stress. Gaining a deeper understanding of the molecular
mechanisms governing A. fumigatus iron acquisition and oxidative stress response may ultimately help to improve the
diagnosis and treatment of invasive aspergillus infections.
Results: This study follows a systems biology approach to investigate how adaptive behaviors emerge from
molecular interactions underlying A. fumigatus iron regulation and oxidative stress response. We construct a Boolean
network model from known interactions and simulate how changes in environmental iron and superoxide levels
affect network dynamics. We propose rules for linking long term model behavior to qualitative estimates of cell
growth and cell death. These rules are used to predict phenotypes of gene deletion strains. The model is validated on
the basis of its ability to reproduce literature data not used in model generation.
Conclusions: The model reproduces gene expression patterns in experimental time course data when A. fumigatus is
switched from a low iron to a high iron environment. In addition, the model is able to accurately represent the
phenotypes of many knockout strains under varying iron and superoxide conditions. Model simulations support the
hypothesis that intracellular iron regulates A. fumigatus transcription factors, SreA and HapX, by a post-translational,
rather than transcriptional, mechanism. Finally, the model predicts that blocking siderophore-mediated iron uptake
reduces resistance to oxidative stress. This indicates that combined targeting of siderophore-mediated iron uptake
and the oxidative stress response network may act synergistically to increase fungal cell killing.
Keywords: Boolean network, Discrete dynamic model, Invasive aspergillosis, Siderophores, Stochastic discrete
dynamical system
Background
Aspergillus fumigatus is a ubiquitous airborne fungus
which has become an increasingly dangerous pathogen
of humans worldwide, causing invasive infections, severe
asthma and sinusitis [1]. The most severe form of A. fumi-
gatus infection, called invasive aspergillosis (IA), occurs
when inhaled A. fumigatus spores germinate into hyphae
and invade lung tissue. IA is a major cause of mortality
in immunocompromised human hosts [2-6]. In immuno-
competent individuals A. fumigatus may trigger allergic
*Correspondence: mbrandon@uchc.edu
1Center for Cell Analysis and Modeling, University of Connecticut Health
Center, 400 Farmington Ave, 06030 Farmington, USA
2Center for Quantitative Medicine, University of Connecticut Health Center,
195 Farmington Ave, 06030 Farmington, USA
Full list of author information is available at the end of the article
reactions and is a major cause of fungal keratitis, an
inflammation of the cornea [7].
Our focus on A. fumigatus oxidative stress response
and iron acquisition is motivated by the following three
arguments. First, several studies show that deletion of
genes involved in either A. fumigatus oxidative stress
response or iron acquisition leads to attenuated virulence
in vivo [5,8-10]. Impairment of the corresponding host
defense mechanisms, e.g. defective ROS production or
inability to sufficiently deplete available iron, also leads
to an increased susceptibility to A. fumigatus infection
[4,10,11]. Second, recent publications present proof of
concept that targeting either A. fumigatus oxidative stress
response or iron acquisition systems may be an effective
treatment strategy [10,12]. Thus oxidative stress response
© 2015 Brandon et al.; licensee BioMed Central. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication
waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise
stated.
Brandon et al. BMC Systems Biology (2015) 9:19
Page 2 of 17
and iron acquisition are important systems contribut-
ing to A. fumigatus pathogenicity, and both systems are
feasible targets for therapeutic intervention. Third, iron
uptake and oxidative stress response networks are known
to interact, and hence more can be learned about the
molecular mechanisms underlying these networks if they
are studied together. In fact, a connection between iron
uptake and oxidative stress response has been described
in both A. fumigatus and S. cerevisiae [13-15]. These
motivating points will now be discussed in greater detail.
Several lines of evidence point to the A. fumigatus ROS-
detoxifying enzymes as key virulence factors and potential
drug targets. Firstly, on the host side, the activation of
the enzymatic complex NADPH oxidase (NOX) and sub-
sequent production of cytotoxic ROS by host phagocytic
cells is a critical mechanism for host defense against fun-
gal pathogens such as A. fumigatus [16-18]. Noteably,
a mouse model of fungal keratitis in the cornea using
mice that do not express a functional NOX complex
showed that neutrophil NOX expression was required
for inhibiting A. fumigatus growth [10]. From a fun-
gal perspective, genes encoding oxidative stress response
enzymes are known to be among the most differentially
expressed genes of A. fumigatus hyphae following expo-
sure to human neutrophils from healthy individuals [19].
Furthermore, A. fumigatus antioxidant enzymes and the
ROS-sensing transcription factor deletion strains show a
heightened sensitivity to ROS in vitro [9,10,20].
Other evidence suggests that the adeptness of A. fumi-
gatus to acquire iron from the host is a major basis of its
pathogenicity. Both the fungus and host require iron for
important cellular functions including respiration, gene
regulation, DNA synthesis, and oxidative stress response
[21]. Iron deprivation of invading pathogens by the host
is a crucial host defense mechanism [22-24]. To combat
this, fungi secrete siderophores, low-molecular-mass iron
binding compounds that sequester iron from host pro-
teins. [25]. A significant body of evidence suggests that
the victor of this battle for iron is a key determinant
of whether infection will persist or be cleared [25-28].
Notably, a mutant A. fumigatus strain unable to produce
both extra- and intracellular siderophores was avirulent
in a mouse model of IA [5]. Any advantage A. fumigatus
has in the battle for iron can be dangerous. For instance,
increased iron in bone marrow is a risk factor for IA in
high-risk patients [11]. Similarly, the heightened suscep-
tibility to fungal infections in neutropenic patients may
be in part due to increased extracellular iron due to the
absence of host cells which mediate iron sequestration
[29].
Leal et al. show that the use of topical drugs to tar-
get either A. fumigatus oxidative stress response or iron
acquisition systems is effective for treating A. fumiga-
tus infection in mice cornea [10,12]. The fungal iron
acquisition system is a particularly promising target
for therapeutic intervention because the fungal proteins
which import ferri-siderophores are one of the few protein
families that are unique to fungi [30]. This might make it
possible to design drugs which specifically target the fun-
gus without affecting the host, perhaps by a “Trojan horse”
approach [31,32]. Furthermore, the iron acquisition and
oxidative stress response networks are connected. Indeed
it was found in A. fumigatus that deletion of a key iron
regulatory protein, sreA, caused increased sensitivity
to superoxide [13]. Also in A. fumigatus deletion of an
intracellular siderophore led to decreased expression of
conidial, but not hyphal, catalase [33]. Similarly, in A.
nidulans oxidative stress was shown to increase the accu-
mulation of an intracellular siderophore [14]. Finally, a
yeast mutant with deletion of genes that regulate the tran-
scription of high-affinity iron transport genes also showed
several phenotypes related to oxidative stress such as
hypersensitivity to hydrogen peroxide [15].
The role of mathematical modeling
The purpose of the present work is to gain a deeper
understanding of the molecular mechanisms underly-
ing the systems that most contribute to A. fumigatus
pathogenicity, the iron acquisition and oxidative stress
response networks. For this purpose, we have constructed
a novel dynamic mathematical model of key molecu-
lar interactions defining these networks. Mathematical
modeling of complex molecular interaction networks
allows for the encoding of dynamic interactions among
molecules, and thus enables the simulation of global net-
work behavior based on information known about indi-
vidual interactions.
Recently, the first computational model of A. fumiga-
tus iron regulation was proposed [34]. Taking a top-down
approach, Linde et al. used gene expression time series
data to reverse engineer a regulatory network and pre-
dict new interactions between transcription factors and
target genes. The authors constructed a system of differ-
ential equations to model changes in gene expression as a
function of other genes in the network. A major challenge
to building differential equations models is that many of
the required parameters are either unknown or unmea-
surable, and so parameters must be estimated by fitting
equations to experimental time series data, which is lim-
ited for A. fumigatus iron regulation and oxidative stress
response.
However, there is a wealth of qualitative data for these
networks, for example the interaction between a tran-
scription factor and a gene, from high-throughput tran-
scriptomic experiments such as microarrays [13,29,35]. In
contrast to the Linde et al. computational study, we take
a bottom-up approach to investigate both iron regula-
tion and oxidative stress response, and we apply a discrete
Brandon et al. BMC Systems Biology (2015) 9:19
Page 3 of 17
dynamic modeling framework. Discrete models make use
of the available qualitative data by encapsulating the reg-
ulatory logic driving a network, and they do not require
kinetic parameters. Simulation of discrete models pro-
vides coarse-grained information as the network evolves
over an arbitrary unit of time in response to broad changes
in some physiological condition. Qualitative observations
generated by these models are extremely useful for inves-
tigating the ability of known or proposed information to
explain current experimental results, studying how per-
turbations may alter global behavior, and for pinpointing
productive future experiments.
Discrete models, in particular Boolean network mod-
els, are routinely used to investigate biological systems
such as gene regulatory networks, signaling pathways,
and metabolic pathways [36-41]. Discrete models have
contributed insights into host-pathogen interactions for
several pathogenic bacteria [42-44]. To our knowledge,
discrete models have not yet been used to study A. fumi-
gatus biology, yet many aspects of yeast biology have
been explored via discrete models [45-47]. This includes
a Boolean network model of metabolic adaptation to
oxygen in relation to iron homeostasis and oxidative
stress [48].
Results and discussion
Description of model species
The model contains an oxidative stress response mod-
ule and a larger iron acquisition module which is made
up of five submodules: siderophore biosynthesis (SB),
iron uptake, iron storage, iron usage, and iron regulation.
Figure 1 is a graphical representation of all model species
(nodes), their interactions (edges), and the sign of the
interaction.
Siderophore biosynthesis
A. fumigatus produces four siderophores, low molecular
mass ferric iron-specific chelators [33]. Two extracellu-
lar siderophores are excreted from the cell to sequester
iron from the extracellular space [8]. And two intracel-
lular siderophores are used for intracellular iron storage
[14,49]. For simplicity, our model considers only one
extracellular siderophore, triacetylfusarinine C (TAFC),
and one intracellular siderophore, ferricrocin (FC), which
have been shown to be the two most abundant and
active A. fumigatus siderophores [50]. The first step in
the biosynthesis of all four siderophores is the hydrox-
ylation of ornithine catalyzed by SidA, an ornithine
monooxygenase.
Iron uptake
Iron uptake is believed to be the main iron homeostasis
control mechanism used by A. fumigatus, in part because
mechanisms of iron excretion have not been found in
fungi [51]. A. fumigatus has three known mechanisms of
iron uptake: low affinity ferrous iron uptake, which has not
yet been characterized at the molecular level, and two high
affinity ferric iron uptake systems, namely siderophore-
mediated iron uptake and reductive iron assimilation
(RIA) [8]. RIA involves the reduction of ferric iron to fer-
rous iron by the ferric reductase FreB and subsequently
the import of ferrous iron by a protein complex consisting
of the ferroxidase FetC and the iron permease FtrA [52].
For simplicity, these three proteins are modeled as a single
species called RIA.
Siderophore-mediated iron uptake is represented in
the model by nodes TAFC, MirB, and EstB. TAFC is
released into the extracellular space to steal ferric iron
from host proteins such as transferrin [53]. A protein fam-
ily called siderophore-iron transporters (SIT) recognizes
and retrieves specific ferri-siderophores. After binding to
Fe3+, the ferri-TAFC complex is taken back up by the
TAFC-specific SIT MirB [54]. After import into the cell
the ferri-TAFC complex is degraded by a TAFC-specific
esterase called EstB [55]. Subsequently, breakdown prod-
ucts are recycled, and iron is released into the cell for
transfer to intracellular siderophores or the iron vacuole
[56].
Iron storage
Unlike bacteria, plants and animals, most fungi lack
ferritin-mediated iron storage [51]. Instead, A. fumigatus
relies on siderophore-mediated iron storage via the intra-
cellular siderophore FC and a siderophore-independent
iron storage unit, the iron vacuole [49,56]. Import of iron
into the vacuole is in part mediated by the protein CccA
which is localized in the vacuolar membrane [56]. The
labile iron pool, a pool of redox-active iron, is also mod-
eled as a transitory state between the release of iron from
ferri-TAFC and the transfer of iron to FC or the vacuole.
Again, since fungi lack mechanisms for iron excretion,
iron storage plays a crucial role in avoiding iron-induced
toxicity. In A. nidulans, FC deficiency was shown to cause
an increase in LIP and a decrease in the oxidative stress
resistance of hyphae [57].
Iron usage
All iron consuming pathways, for example heme biosyn-
thesis, TCA cycle, respiration, and ribosome biogenesis,
are modeled as a single species named ICP.
Regulation
Iron is toxic in excess; thus tight regulatory mechanisms
are required to maintain iron homeostasis. Iron regulation
in A. fumigatus is controlled by two central transcrip-
tion factors: the bZip CCAAT-binding transcription fac-
tor HapX and the GATA transcription factor SreA [13,29].
HapX and SreA are postulated to sense intracellular iron
Brandon et al. BMC Systems Biology (2015) 9:19
Page 4 of 17
Iron Regulation
Iron
Storage
Iron Usage
Oxidative Stress
Response
Iron Uptake
Siderophore
Biosynthesis
sreA
hapX
SreA
HapX
RIA
EstB
MirB
TAFC
SidA
LIP
VAC
thioredoxin
pathway
Fe3+
CccA
O2
-
ROS
SOD2/3
Yap1
Cat1/2
ICP
FC-Fe
FC+Fe
Figure 1 Model interaction diagram of A. fumigatus iron regulation and oxidative stress response. Rectangles represent genes. Ovals represent
other molecules. Fe3+ and O−
2 are external parameters to describe the physiological state of a fungal cell. A →B represents activation. A ⊣B
represents inhibition.
levels through a posttranslational mechanism similar to
the mechanism employed by a closely related species,
the fission yeast Schizosaccharomyces pombe [27]. In S.
pombe, orthologs of HapX and SreA physically interact
with a monothiol glutaredoxin Grx4 which is localized
along the nuclear rim [58-60]. When intracellular iron lev-
els are low Grx4 maintains SreA in an inactive state [59].
When intracellular iron levels are high, Grx4 inactivates
HapX by directing its export from the nucleus [58]. Hence,
intracellular iron blocks HapX function while activating
SreA function at the posttranslational level. Furthermore,
SreA represses transcription of hapX when intracellular
iron levels are high, while HapX represses transcription
of sreA when intracellular iron levels are low. Both tran-
scriptional and posttranslational regulatory mechanisms
are modeled.
SreA transcriptionally represses genes coding for pro-
teins involved in iron uptake, including sidA, mirB,
estB, and those involved in RIA [13]. HapX activates
siderophore biosynthesis, in part by upregulating the pro-
duction of the precursor ornithine, and activates the
transcription of mirB [29]. HapX indirectly activates the
transcription of sidA, estB, and the genes involved in
RIA through its repression of sreA. Additionally, HapX
represses iron consuming pathways, cat1, and cccA at the
transcriptional level.
Oxidative stress response
NOX expressed by host phagocytic cells catalyzes the con-
version of oxygen to the the extremely reactive superox-
ide anion, O−
2 . Contact between neutrophils and hyphae
triggers a respiratory burst, the targeted release of O−
2
from the neutrophil into the extracellular space where it
diffuses into nearby hyphal cells. The A. fumigatus ROS-
sensing transcription factor Yap1 is believed to be the
main regulator of antioxidant defense against O−
2 and
hydrogen peroxide, H202 [61,62]. Yap1 typically resides in
the cytoplasm, yet under oxidative stress conditions Yap1
localizes to the nucleus and from there controls, directly
or indirectly, the expression of key ROS-detoxifying
enzymes including superoxide dismutases (SODs), cata-
lases, and thioredoxin peroxidases (peroxiredoxins) [61].
Elevated free iron levels (high LIP) in the cell also con-
tribute to the formation of ROS [63].
SODs catalyze the conversion of O−
2
to less reac-
tive H2O2 which can then be converted to non-reactive
Brandon et al. BMC Systems Biology (2015) 9:19
Page 5 of 17
H2O by either catalases or peroxiredoxins. A. fumigatus
produces four SODs, yet only the mitochondrial SOD2
and cytoplasmic SOD3 are modeled here since both are
most strongly expressed in hyphae, the tissue invasive
form of this pathogen, as opposed to in conidia [20]. A.
fumigatus hyphae produce two catalases, Cat1 and Cat2,
which break down hydrogen peroxide [9]. The thiore-
doxin pathway in A. fumigatus is not well characterized;
however, two putative peroxiredoxins and five putative
thioredoxins have been identified [10,61]. Briefly, per-
oxiredoxins reduce H202 and by doing so become oxi-
dized, a non-functional state. Thioredoxins then reduce
the oxidized peroxiredoxins back to their functional state
so that more H202 can be reduced [64]. In the model
the thioredoxin pathway is modeled as a single vari-
able. Note that in Figure 1 the ROS species has a self-
activating arrow. The purpose of this interaction is to
enforce “memory” in the system, i.e. if ROS is high
at the current time step and antioxidant enzymes are
not expressed or inactive, then the ROS variable should
“remember” to remain high until antioxidant enzymes are
active.
Building and simulating the mathematical model
The model presented in this paper is discrete. This means
species can take on only a finite number of states, and
the state of each species is iteratively updated at discrete
time steps according to logical rules. The discrete model
presented here is a Boolean network model, meaning
that each species can take on only two states (e.g. low
expressed or high expressed; low active or high active),
which may be represented numerically by either a 0 or
a 1. Furthermore, the rules determining how species are
updated are Boolean functions.
We conducted an extensive literature survey to iden-
tify key species involved in the A. fumigatus iron reg-
ulatory and oxidative stress response networks as well
as the interactions of each species with other species
in the networks (Figure 1). Table 1 gives a biological
description of each species and the meaning assigned to
its states. Note that for different species we may assign
different meanings to their states. Importantly, the two
species iron and superoxide should be thought of as
external parameters since they are meant to distinguish
between different physiological conditions that are reflec-
tive of the host-pathogen interaction. Iron and superox-
ide have no regulators (incoming arrows) (see Figure 1)
and so, unlike other species, a fixed state is chosen at
the start of a simulation and this state will never be
updated.
Next we integrated all identified interactions into a
dynamic framework by specifying, through logical rules
called update rules, how each species transitions between
its two states based upon the states of its inputs. Table 2
Table 1 List of species, their biological type, and their
model states
Species
Type
Model states
0
1
hapX
Gene
Low expressed
High expressed
sreA
Gene
Low expressed
High expressed
HapX
Protein; bZip
CCAAT-binding TF
Low active
High active
SreA
Protein; GATA TF
Low active
High active
RIA
Enzyme complex;
reductive iron
assimilation
Low active
High active
EstB
Enzyme;
TAFC-specific
esterase
Low active
High active
MirB
Protein;
TAFC-specific
importer
Low active
High active
SidA
Enzyme; ornithine
monooxygenase
Low active
High active
TAFC
Extracellular
siderophore
Low synthesized
High synthesized
ICP
Iron consuming
pathways
Low active
high active
LIP
Labile iron pool
Low iron
High iron
CccA
Protein; iron
importer to
vacuole
Low active
High active
FC+Fe
Intracellular
siderophore w/
bound iron
Low iron
High iron
FC−Fe
Intracellular
siderophore w/o
bound iron
Low synthesized
High synthesized
VAC
Vacuole
Low iron
High iron
ROS
Reactive oxygen
species
Low ROS
High ROS
Yap1
Protein; bZip TF
Low active
High active
SOD2/3
Enzyme;
superoxide
dismutase
Low active
High active
Cat1/2
Enzymes; hyphal
catalases
Low active
High active
Thioredoxin
P.
Enzyme pathway
Low active
High active
Iron
Physiological state
Low iron
High iron
Superoxide
Physiological state
Low superoxide
High superoxide
lists the update rule for each species as a Boolean function
along with a summary of experimental support for each
rule. The model is available in SMBL qual format, a stan-
dard language for representation of qualitative models of
biological networks [65], see Additional file 1.
Brandon et al. BMC Systems Biology (2015) 9:19
Page 6 of 17
Table 2 Update rules of model species and supporting literature citations
Update rules
Literature support
1 hapX(t+1) = NOT SreA
Transcription of hapX is repressed by SreA [13,29].
2 sreA(t+1) = NOT HapX
Transcription of sreA is repressed by HapX [13,29].
3 HapX(t+1) = hapX AND (NOT LIP)
An ortholog of HapX is inactivated by intracellular iron [58].
4 SreA(t+1) = sreA AND LIP
An ortholog of SreA is activated by intracellular iron [59,60].
5 RIA(t+1) = NOT SreA
SreA transcriptionally represses RIA genes [13].
6 EstB(t+1) = NOT SreA
SreA transcriptionally represses estB [13].
7 MirB(t+1) = HapX AND (NOT SreA)
HapX transcriptionally activates mirB [29]. SreA transcriptionally represses
mirB [13].
8 SidA(t+1) = HapX AND (NOT SreA)
HapX up regulates the SidA substrate ornithine [29]. SreA transcriptionally
represses sidA [13].
9 TAFC(t+1) = SidA
SidA catalyzes the first step in siderophore biosynthesis [5,8]
10 ICP(t+1) = (NOT HapX) AND (VAC OR FC+Fe)
HapX represses consumption of intracellular iron [29].
11 LIP(t+1) = (TAFC AND MirB AND EstB) OR (Iron AND RIA)
TAFC sequesters iron from the extracellular space [8]. MirB imports ferri-
TAFC [54]. EstB
degrades ferri-TAFC bonds and releases free iron [55]. RIA compensates for
a lack of
siderophores when grown in high iron media [33].
12 CccA(t+1) = NOT HapX
HapX transcriptionally represses cccA [29].
13 FC−Fe(t+1) = SidA
SidA catalyzes the first step in siderophore biosynthesis [5,8]
14 FC+Fe(t+1) = LIP AND FC−Fe
FC is involved in intracellular iron storage [14,49].
15 VAC(t+1) = LIP AND CccA
CccA mediates import of intracellular iron into the vacuole [56].
16 ROS(t+1) = LIP OR
Elevated free iron levels catalyze the formation of ROS [63].
Superoxide AND
NOT (SOD3 AND ThP AND Cat1/2)
OR
SODs convert O−
2 to H2O2 [20]. Either catalases or thioredoxin
ROS AND
NOT
SOD3 AND (ThP OR Cat1/2)
convert H2O2 to non-reactive H2O [9,64].
17 Yap1(t+1) = ROS
Yap-1 is activated by superoxide [61,62].
18 SOD2/3(t+1) = Yap1
Yap-1 activates transcription of sod2/3 [61].
19 Cat1/2(t+1) = Yap1 AND (NOT HapX)
Yap-1 activates transcription of cat1/2 [61]. HapX transcriptionally represses
cat1 [29].
20 ThP(t+1) = Yap1
Yap-1 activates transcription of thioredoxin peroxidases [61].
21 Iron(t+1) = Iron
External parameter.
22 Superoxide(t+1) = Superoxide [= NOT Superoxide, Figure 5 only]
External parameter.
Species that appear on the right side of the = represent states at time t.
In general, the dynamic behavior of discrete models
is simulated by starting from an initial state and then
enumerating the changing state space as each species is
updated over a specified number of iterations called time
steps. The result of deterministic simulations, when all
species are updated simultaneously at each time step, is
shown in Figure 2. This system has no steady state solu-
tion for any of the four external conditions. All long term
behavior is oscillatory, i.e. the stable states form a limit
cycle and 100% of the 1048576 states converge to the limit
cycle displayed.
Many biological processes such as gene expression have
been found to exhibit a high degree of stochasticity
[66-69]. Furthermore, protein levels can differ signif-
icantly among cells in a population [70,71]. To our
knowledge no single cell gene expression or protein level
measurements are available for A. fumigatus. Hence in
order to make comparisons to experimental data possible,
we needed to account for the variability that one observes
in a population of cells. We accounted for this variabil-
ity by simulating randomness in the update of species.
At each time step, rather than updating all species, some
species are randomly selected to be updated, while the
unselected species are left in their current state. We
assume that the average of many of these stochastic simu-
lations represents a population level measurement.
Brandon et al. BMC Systems Biology (2015) 9:19
Page 7 of 17
State:
Low High
Low
Fe3+
High
Fe3+
Extracellular
Fe3+ & O2
-
Levels
hapX
sreA
HapX
SreA
RIA
EstB
MirB
SidA
TAFC
ICP
LIP
CccA
FC-Fe
FC+Fe
VAC
ROS
Yap1
SOD2/3
Cat1/2
ThP
O
w
o
L
2
-
Low
Fe3+
High
Fe3+
O
h
gi
H
2
-
Wildtype
Figure 2 Stable states of A. fumigatus iron regulatory and oxidative stress response networks. This figure shows the cyclic attractor for each of the
four possible external conditions. States transition from top to bottom. Under both low iron conditions ICP is in state 0 (low) the majority of the
cycle. Under both high superoxide conditions ROS is in state 1 (high) the majority of the cycle.
Linking model simulation results to phenotype predictions
For the results presented in this paper, we ran 100 inde-
pendent stochastic simulations (initialized in the same
state) and, for each species, calculated the average state
at each time step. From there, we counted the number
of times a species took on any average state throughout
the simulation period. We can plot a histogram of these
counts to visualize a distribution of species’ average state
across 100 simulations, as in Figure 3. To characterize
long-term behavior, we introduce a measure called the sta-
ble distribution mean (SDM) for a given species under
a given set of initial conditions. The SDM is simply the
mean of the distribution of the average states from time
steps 100 to 200. Excluding the first 100 time steps from
the calculation gives the model time to settle into a stable
configuration.
We first simulated the Boolean network model of wild
type A. fumigatus under each of the four possible condi-
tions: (1) low iron and low superoxide, (2) high iron and
low superoxide, (3) low iron and high superoxide, and (4)
high iron and high superoxide. Figure 3 (A) - (C) show the
distributions of average states across 100 wild type sim-
ulations for six selected species under three of the four
conditions. Wild type distributions are not shown for the
remaining condition; instead Figure 4(B) and (D) show
trajectories, the average state at each time step, for eight
selected species.
For both low iron conditions, we observed that HapX,
the transcription factor activating iron uptake and
repressing iron consumption, is more active than SreA,
the transcription factor repressing iron acquisition. This
leads to strong activity of proteins related to siderophore-
mediated iron uptake (MirB) and reductive iron assim-
ilation (RIA). Conversely, for the high iron conditions
we observe that SreA is more active than HapX. Conse-
quently, activity of both MirB and RIA are significantly
reduced as compared to the low iron, low superoxide
condition. These results recapitulate experimental obser-
vations [29,72].
ROS-detoxifying enzymes, SOD2/3 and Cat1/2, are
moderately active in the low iron, low superoxide con-
dition. As expected, since free iron and superoxide con-
tribute to ROS, the activity of SOD2/3 and Cat1/2 are
elevated in high iron and high superoxide conditions. Fur-
ther, we observed that in low iron conditions Cat1/2 is less
active than SOD2/3. This makes sense because catalases
Brandon et al. BMC Systems Biology (2015) 9:19
Page 8 of 17
Low growth
High growth
Minimal growth
Overwhelming cell death
High cell death
Low cell death
A
B
C
D
0
5
10
Count
Low Fe3+, High O2
-
High Fe3+, High O2
-
Phenotype Reference
Cat1/2
HapX
MirB
RIA
SOD2/3
SreA
Variable
State
State
Count
Count
10
5
0
15
10
5
0
0.0
0.25
0.5
0.75
1.0
0.0
0.25
0.5
0.75
1.0
0.0
0.25
0.5
0.75
1.0
State
Condition
O2
-=0, Fe3+=0
Average state
0.0
0.25
0.5
0.75
1.0
O2
-=0, Fe3+=1
O2
-=1, Fe3+=0
O2
-=1, Fe3+=1
ICP
ROS
Variable
Low Fe3+, Low O2
-
Figure 3 Summary of model wild type phenotypes. (A)-(C) Histogram of average states of six species from time steps 100-200 (i.e., the model
reaches a stable configuration before counting begins). Vertical dashed lines mark stable distribution means (SDM). (D) The SDM of ICP and ROS for
a wild type fungal cell under each of the four conditions overlayed with a depiction of the phenotype reference. If the SDM of ICP is 0, then we
interpret the model observation as minimal or no growth. An ICP SDM in (0, 0.33) is interpreted as low growth. Otherwise, an ICP in [0.33, 1] signifies
a high growth phenotype. If the ROS SDM falls in [0.66,1] we interpret this as high cell death. When the SDM of ROS is 1, we assume the ROS is so
overwhelming that the entire population dies. Otherwise, for an ROS SDM in [0, 0.66) the interpretation is low cell death.
require heme as a cofactor whereas SODs instead require
copper, zinc or manganese [20].
Based on experimental results of wild type A. fumiga-
tus growth under each of the four conditions [8,20,33],
we used the stable distribution mean (SDM) of model
variables ROS and ICP to establish a phenotype refer-
ence according to the following rules (see Figure 3). If the
SDM of ICP is 0, then we interpret the model observa-
tion as minimal or no growth. An ICP SDM in (0, 0.33) is
interpreted as low growth. Otherwise, an ICP in [0.33, 1]
signifies a high growth phenotype. If the ROS SDM falls
in [0.66,1] we interpret this as high cell death. When the
SDM of ROS is 1, we assume ROS is so overwhelming that
the entire population dies. Otherwise, for an ROS SDM in
[0, 0.66) the interpretation is low cell death. These rules
bin wild type model behavior to match what we observe
experimentally. We then used this set of rules to infer the
severity of model knockouts.
Stochastic simulations reproduce in vitro time course data
We validated the model on the basis of its ability to repro-
duce transcriptional time course data from a previously
published study and data generated in this study. In both
experiments, A. fumigatus is grown in iron depleted min-
imal media (low iron, low superoxide conditions). After
an incubation period, iron is added to the media (high
iron, low superoxide conditions) and gene expression is
measured over a period of hours either using microarrays
(Schrettl et al., 2008 [13]) or by qRT-PCR (this study, see
Methods).
To mimic the switch from low iron to high iron con-
ditions, all simulations were initialized from a state of
iron starvation (Figure 4G). Iron and superoxide were
fixed at 1 and 0, respectively, throughout model simula-
tions. Experimental results are displayed alongside model
simulation results in Figure 4. From the Schrettl et al.
study, we plot all time course data for genes which corre-
spond to species in the model. For knockout simulations,
the state of the corresponding species is fixed at 0. To
distinguish between model and experimental knockouts,
using sreA as an example, we write sreA=0 to refer to
model knockouts and sreA to refer to experimental
knockouts.
The model provides a good qualitative reproduction of
changes in gene expression over time. Additionally the
model captures the relative differences in degrees of up-
Brandon et al. BMC Systems Biology (2015) 9:19
Page 9 of 17
Figure 4 Model simulation results and experimental time course data following a switch from low iron, low superoxide to high iron, low
superoxide conditions. (A) Gene expression from a qRT-PCR experiment conducted in this study. (C), (E) Gene expression from a microarray
experiment by Schrettl et al., 2008 for a wild type and sreA strain, respectively [13]. (B), (D), (F), (H) Simulated trajectories for corresponding model
species plotted as the average state at each time step across 100 stochastic simulations. (G) All simulations were initialized from this state
representing iron starvation. In (H) trajectories are generated by a model with post-translational regulation of HapX and SreA by iron (PTL) and an
altered model with transcriptional regulation of hapX and sreA by iron (TS).
Brandon et al. BMC Systems Biology (2015) 9:19
Page 10 of 17
or down-regulation among genes. For ease of exposition,
we discuss the following results in the syntax of model
species even though some model species refer to amount
of protein while the experimental results refer to amount
of transcript.
Wild type results
For wild type A. fumigatus, the experimental and model
simulation results show the same expression patterns.
Following the switch from low to high iron, catalases
Cat1/2, the vacuolar iron importer CccA, and sreA were
quickly up-regulated, then slowly decreased and leveled
off. After the addition of iron, the expression of hapX,
siderophore biosynthesis enzyme SidA, ferri-siderophore
importer MirB, ferri-siderophore esterase EstB, and
reductive iron assimilation RIA (ftrA in the experimen-
tal data) were quickly down-regulated and remained
low. Moreover, the model recapitulates experimental
observations that among species contributing to iron
uptake, SidA and MirB were less active under high iron
conditions, as compared to the activity of RIA and EstB.
sreA results
Experimental and model simulation results are also in
agreement for the sreA deletion strain, except for SidA
and MirB which we discuss shortly. As in the wild type
case, expression of CccA and Cat1/2 increased sharply
after the addition of iron. However in the sreA knock-
out, expression of CccA and Cat1/2 remained high over
time. This can be attributed to the fact that iron uptake
mechanisms were depressed in a sreA mutant. Indeed,
in contrast to the wild type case, both experimental and
model simulation results showed no change in the expres-
sion of hapX, EstB, or RIA despite being exposed to high
iron over a long period of time.
In the sreA deletion experimental data the amount of
sidA transcript remained the same, whereas the amount
of SidA enzymatic activity in the model simulation plum-
meted to very low. This in fact is not a discrepancy
and serves to illustrate an important point. Although
both HapX and SreA ultimately activate and respectively
repress SidA enzymatic activity, only SreA directly tran-
scriptionally regulates sidA [13,29]. Instead, HapX up-
regulates the production of ornithine, the SidA substrate.
This explains the derepression of sidA in the experimental
data yet the lack of SidA activity in the model simula-
tion. A difference between amount of sidA transcript and
SidA enzymatic activity is not visible in the wild type
data because HapX indirectly regulates the transcription
of sidA through repression of sreA; this feature is lost in a
sreA mutant.
The discrepancy between gene expression of mirB
in the experiment and activity of MirB in the model
is unexpected since MirB is known to be regulated
transcriptionally by both HapX and SreA [13,29]. This
may suggest that MirB is in fact not regulated transcrip-
tionally by HapX. Or alternatively, since MirB is known
to transport other siderophores it may have additional
regulators [54].
Regulation of HapX and SreA by iron
The post-translational regulation of S. pombe orthologs of
HapX and SreA by iron has been investigated [58-60]. A.
fumigatus HapX and SreA are postulated to sense intra-
cellular iron levels through a similar post-translational
mechanism, but the corresponding mechanism has not
yet been identified [27]. We included in the model both
the known transcriptional regulation of hapX and sreA,
by SreA and HapX respectively, and the proposed but
not yet verified post-translational regulation of HapX and
SreA by intracellular iron. Additionally, we analyzed a
modified model whereby hapX and sreA were regulated
transcriptionally by iron, and all other interactions are
the same. Both versions of the model were consistent
with gene expression data for the wild type. However,
the model with HapX and SreA regulated by iron at
the post-transtlational level, but not the modified model,
agreed with the Schrettl et al. hapX gene expression data
for the sreA mutant strain (see Figure 4H). This pro-
vides support for the hypothesis that, as in S. pombe,
a post-translational regulation of the iron regulatory
proteins HapX and SreA by iron is in fact employed by
A. fumigatus.
Model knockout simulations recapitulate experimental
gene deletion results.
Next, we systematically analyzed the effect of all single and
double knockouts on model predicted phenotypes under
each of the four external conditions. Key observations
from wild type and knockout simulations are summarized
in Table 3.
Iron regulation knockouts
For the low iron conditions, the SidA = 0 knockout led
to minimal or no growth (SDM of ICP = 0). However, no
growth defects were observed when RIA = 0 under the
same conditions. In high iron conditions, the SidA = 0
knockout did not deviate from the wild type high growth
phenotype. However, in high iron conditions, a double
RIA = SidA = 0 knockout led to minimal or no growth.
These results are consistent with experimental results
showing: (1) a sidA but not ftrA mutant is avirulent in
a mouse model of aspergillosis [5,8], and (2) that RIA can
compensate for a lack of siderophores in high iron but not
low iron conditions [33].
Also in agreement with experiments, under low iron
conditions the TAFC = 0 knockout led to more severe
growth defects than the FC−Fe =0 knockout [33]. Knocking
Brandon et al. BMC Systems Biology (2015) 9:19
Page 11 of 17
Table 3 Summary of observations from model wild type and knockout simulations
Condition
Strain
Long term model behavior
Cell
Cell
Support
Conflicts
ICP
ROS
Other interesting behavior
Growth
Death
O−
2 = 0
wt
0.29
0.44
• TAFC = FC−Fe = 0.52; high siderophore production
−
−
[5,8]
• Cat1/2 = 0.29; SOD2/3 = ThP = 0.44
prediction
• FC−Fe/FC+Fe =1.7
[14]
Fe3+ = 0
hapX = 0
0
0
• SidA = 0
−
−
[29]
SidA = 0 or TAFC = 0
0
0
−
−
[5,8,33,52]
FC−Fe = 0
0.11
0.43
−
−
[14,33]
EstB = 0 or MirB = 0
0
0
• TAFC = FC−Fe = 1; accumulation of siderophores
−
−
[55]
Yap1 = 0 or SOD2/3 = 0
0.29
1
−
+
[10,20]
O−
2 = 0
wt
0.48
0.62
• TAFC = FC = 0.18; low siderophore production
+
−
[5,8]
• Cat1/2 = 0.54, SOD2/3 = ThP = 0.61
[13]
• FC−Fe/FC+Fe = 1
[14]
Fe3+ = 1
sreA = 0
1
1
• Derepression of hapX, RIA, CccA, & Cat1/2
+
+
[13,52]
• LIP = VAC = 1; iron overload
[13]
SidA = 0
0.42
0.61
+
−
[5,8,33]
RIA = 0
0.29
0.45
• SidA = 0.52; increased siderophore production
−
−
[8,33]
SidA = RIA = 0
0
0
−
−
[8,33]
Yap1 = 0 or SOD2/3 = 0
0.49
1
• Decreased resistance to Fe3+
+
+
[10,20]
O−
2 = 1
wt
0.28
0.78
• SOD2/3 = ThP = 0.76; Cat1/2 = 0.40
−
+
[20,33,61]
hapX = 0
0
0.58
• Derepressed Cat1/2 and increased resistance to O−
2
−
−
prediction
Fe3+ = 0
SidA = 0, TAFC = 0,
0
1
• Decreased resistance to O−
2
−
+
prediction
MirB = 0 or EstB = 0
Yap1 = 0, SOD2/3 = 0,
0.29
1
• Decreased resistance to O−
2
−
+
[9,10,20,61]
Cat1/2 = 0 or ThP = 0
O−
2 = 1
wt
0.49
0.73
• FC−Fe/FC+Fe = 1
+
+
[14,20,33]
• SOD2/3 = ThP = 0.73; Cat1/2 = 0.63
prediction
Fe3+ = 1
sreA = 0
1
1
• Decreased resistance to Fe3+
+
+
[13]
Yap1 = 0, SOD2/3 = 0,
0.47
1
• Decreased resistance to Fe3+ and O−
2
+
+
[9,20,61]
Cat1/2 = 0 or ThP = 0
Numerical values in the ‘Long Term Model Behavior’ column represent SDMs. A −denotes low cell growth or low cell death, while a + denotes high cell growth or
high cell death. Citations for supporting and conflicting literature are provided.
out any part of the siderophore iron uptake system under
low iron conditions (TAFC = 0, MirB = 0, or EstB = 0)
resulted in a minimal growth phenotype. Interestingly,
we observed an accumulation of siderophores for either
MirB = 0 or EstB = 0 under low iron conditions, a behav-
ior which has been observed experimentally in an estB
mutant [55].
The hapX = 0 knockout displayed a minimal growth
phenotype under low iron conditions, but had no defects
under high iron conditions. Conversely, the sreA = 0
knockout led to iron overload (SDM of LIP = 1) and cell
death by overwhelming ROS (SDM of ROS = 1) under
high iron conditions, but had no defects under low iron
conditions. This recapitulates experimental results show-
ing that growth defects of a hapX mutant are confined
to low iron conditions while growth defects of a sreA
mutant are confined to high iron conditions [13,29].
Oxidative stress response knockouts
As expected, wild type ROS-detoxifying enzyme activity
was lowest under the low iron, low superoxide condition.
The SDM of Cat1/2 was less than that of SOD2/3 and
the thioredoxin pathway (abbreviated to ThP in Table 3)
whenever iron was low. Under low superoxide conditions
the model predicted a Yap1 = 0 or SOD2/3 = 0 knockout,
but not a Cat1/2 = 0 or Thp = 0 knockout, to be fatal.
This observation demonstrates that the model accounts
for the redundancy that both catalases and peroxiredoxins
reduce H2O2. Under high superoxide conditions, model
knockouts of any of the four oxidative stress-related
Brandon et al. BMC Systems Biology (2015) 9:19
Page 12 of 17
species had a high cell death phenotype due to over-
whelming ROS.
The observation that Yap1 or SOD2/3 deletion is
more severe than deletion of Cat1/2 or blocking of the
thioredoxin pathway is consistent with experimental
results from Leal et al. which show that yap1 and
sod1/2/3 mutant strains are sensitive to neutrophil-
mediated oxidative stress, whereas a cat1/2 strain is
not [10]. Yet the observation is inconsistent with a result
from the same study which showed that blocking the
thioredoxin pathway results in a reduction of in vivo
hyphal growth similar to deletion of either yap1 or sod2/3
[10]. An earlier study, which demonstrated that a cat1/2
mutant showed increased sensitivity to H2O2 in vitro
and delayed growth during infection in a rat model of
aspergillosis, further conflicts with the Leal et al. study
and provides support for the model predicted phenotype
of a Cat1/2 = 0 knockout [9]. Overall, the severity of
experimental knockout results seem to be exaggerated
by some model predictions. In particular, deletion of
either yap1 or sod2/3 should not result in high cell
death in the absence of oxidative stress, yet under low
superoxide conditions the model predicted knockouts a
high cell death phenotype for Yap1 = 0 and SOD2/3 = 0
knockouts.
Discrepancies between experimental results and model
predictions indicate that important species or interactions
may be missing from the model. This may reflect that our
understanding of A. fumigatus oxidative stress response
is still not complete. For instance, there may be uniden-
tified redundancy, some of which could be attributed
to LaeA-controlled secondary metabolites which inhibit
neutrophil production of NOX [73]. Yet knockout exper-
iments of several of these metabolites as well as laeA
suggest they may play no role in protecting A. fumiga-
tus from neutrophil-mediated oxidative stress [10]. Since
model-predicted phenotypes of Yap1 and Yap1-regulated
species knockouts are most overstated in low superox-
ide conditions, it is possible that the model lacks some
constitutively active or baseline antioxidants which may
be useful for neutralizing ROS produced during normal
cellular activities but may not necessarily be helpful in
combating oxidative stress. As more research is done to
characterize new players in the A. fumigatus oxidative
stress response network, the oxidative stress response
module of this model can be improved and new insights
may be gained.
Model suggests combined blocking of iron uptake and
oxidative stress response
Of the four conditions in Table 3, the low iron and high
superoxide condition most resembles the environment
that A. fumigatus cells experience inside a mammalian
host. Under this condition, model knockouts which
impaire siderophore-mediated iron uptake increased A.
fumigatus sensitivity to oxidative stress. A similar rela-
tionship between high-affinity iron uptake and sensitivity
to oxidative stress has been observed in yeast [15]. This
observation led us to wonder what the model would pre-
dict for ROS levels if combined blocking of siderophore-
mediated iron uptake and oxidative stress response were
simulated under less rigid external conditions.
Several experimental studies have investigated target-
ing either siderophore-mediated iron uptake or oxidative
Sample O2- Trajectories
Time steps
0
1
0
10
20
30
40
50
0
1
State
State
ROS Stable Distributions
State
Count
0
5
10
0.0
0.25
0.5
0.75
1.0
Treatment
Both
Drug 1
Drug 2
No Drug
A
B
Figure 5 Model results comparing the application of two hypothetical anti-fungal drugs to treat a simulated A. fumigatus infection. Iron is fixed at
low (0) while superoxide is allowed to randomly toggle between low (0) and high (1). (A) Two representative superoxide trajectories. (B) ROS stable
distributions for no drug, either drug individually, or both drugs together. Vertical dashed lines represent ROS SDMs. Drug 1 targets siderophore-
mediated iron uptake. Drug 2 targets oxidative stress response. Simulations are initialized from the state in row 2 of the low iron, high superoxide
block in Figure 2.
Brandon et al. BMC Systems Biology (2015) 9:19
Page 13 of 17
stress response, but none have investigated any potential
therapeutic gain by combined targeting. TAFC and MirB
are promising drug targets since they are easily acces-
sible and unique to fungi [30]. Leal et al. demonstrate
proof of concept that using lipocalin-1 to sequester TAFC
improves the treatment of topical A. fumigatus infection
[12]. An anti-cancer drug, PX-12, which is known to block
the thioredoxin pathway also shows promise as an anti-A.
fumigatus drug [10].
To investigate the effects of simultaneously inhibiting
siderophore-mediated iron uptake and oxidative stress
response, we simulated treatment with two hypothetical
drugs loosely based on lipocalin-1 and PX-12. Hypotheti-
cal drug 1 binds and inactivates TAFC from 50% of fungal
cells. Hypothetical drug 2 blocks the thioredoxin pathway
in 50% of fungal cells. For these simulations, we held
iron fixed at 0 but allowed superoxide to randomly toggle
between 0 and 1. This setup recapitulates host defense
mechanisms of sustained iron witholding and intermittent
respiratory bursts. Figure 5(A) shows two representative
superoxide trajectories when random toggling is allowed.
Figure 5(B) shows ROS stable distributions from the aver-
age of 100 stochastic simulations for treatment with Drug
1, Drug 2, both drugs or neither drug. To simulate the drug
treatments with 50% efficacy, we fixed either TAFC, ThP,
or both at 0 for 50 of the 100 simulations. Drug 1 alone
has no effect, Drug 2 alone increases the ROS SDM from
0.64 to 0.74, and the combination of Drug 1 and Drug 2
further increases the ROS SDM to 0.83. This result sug-
gests that combined targeting of siderophore-mediated
iron uptake and the oxidative stress response network may
act synergistically to increase fungal cell killing.
Conclusions
In this study we introduce a stochastic Boolean model
of the iron regulatory and oxidative stress response net-
works in A. fumigatus. Model simulations of a population
of A. fumigatus cells reproduces gene expression patterns
in experimental time course data when A. fumigatus is
switched from a low iron to a high iron environment. In
addition, the model is able to accurately represent the phe-
notypes of many knockout strains under varying iron and
superoxide conditions.
We drew three main observations from model analysis.
First, the model provides support for the hypothesis that
A. fumigatus iron regulatory proteins, HapX and SreA, are
regulated by iron at the post-translational level. Second,
based on discrepancies between model knockout sim-
ulations and experimental observations of A. fumigatus
oxidative stress response related mutants, it is likely that
important enzymes or pathways involved in A. fumigatus
ROS-detoxification remain uncharacterized. And third,
impairment of siderophore-mediated iron uptake mecha-
nisms reduces A. fumigatus resistance to oxidative stress.
This fact could be exploited when designing a treatment
strategy.
Deterministic simulation (as well as individual stochas-
tic simulations, data not shown) of the model predicts
sustained oscillations under each of the four external con-
ditions (Figure 2). On the other hand when a population
of cells is modeled by averaging many stochastic simula-
tions, the model converges to a steady state distribution
(Figure 3). Without single cell data, it is unclear how to
interpret discrepancies between an individual (determin-
istic or stochastic) simulation and the average of many
stochastic simulations. It is conceivable that, in agree-
ment with individual model simulations, expression of
iron homeostasis and oxidative stress response genes in a
single A. fumgatus cell may continually oscillate, perhaps
always overshooting and undershooting ideal intracellu-
lar iron levels. A recent study in E coli reported damped
oscillations in the expression of genes involved in iron
homeostasis in a single E. coli cell undergoing a switch
from high iron to low iron conditions [21]. Future single
cell experiments of A. fumigatus could shed light on how
stochasticity arises in fungal iron regulatory and oxidative
stress response networks.
The intended future application of this model is to
incorporate it into a multi-scale systems biology model of
invasive aspergillosis in the lung. The ultimate goal of the
proposed multi-scale model is to capture the effect of the
initial inoculum on disease outcome and to allow for the
investigation of a variety of therapeutic interventions.
Methods
Computational methods
Discrete modeling framework
In order to translate the network interactions depicted in
the diagram of Figure 1 into a dynamic discrete model,
namely a time- and state-discrete dynamical system, the
state transitions for each species must be specified by
assigning an update rule that describes how the species’s
state will be updated at the next time step based upon the
states of its inputs at the current time step. Since this is a
Boolean model, each species can take on only two states
and update rules are Boolean functions. It is convenient
to encode update rules in an object called a transition
table. As an example, consider the iron-sensing transcrip-
tion factor SreA. From the literature we know that when
intracellular iron levels are low, SreA is kept in an inac-
tive state [59,60]. Based on this we decide the update of
SreA should depend on two inputs, its gene sreA and the
labile iron pool LIP. These interactions are represented in
Figure 1 as the two edges incident on SreA. Based on the
state descriptions assigned to SreA, sreA, and LIP listed in
Table 1, we obtain the following table which determines
which state SreA will transition to at time step t + 1 based
on the states of sreA and LIP at the current time step, t.
Brandon et al. BMC Systems Biology (2015) 9:19
Page 14 of 17
Using Table 1 to translate the state descriptions into 0’s
and 1’s, we obtain the following transition table.
This table encodes the AND function. Both sreA AND
LIP must be in state 1 at the time t for SreA to be in state
1 at time t + 1. Any Boolean function can be written using
only AND, OR, and NOT gates (see Table 2).
For ease of computation, we prefer to work with a math-
ematical object rather than transition tables or Boolean
functions. Any discrete dynamical system can be rep-
resented as a system of polynomial equations over a
finite field. A model in this form is called a polynomial
dynamical system (PDS) and can be analyzed using theory
and tools from computational algebra [74,75]. Since each
species in a Boolean network model can take on only two
states, the finite field for our model is F2 ∼= Z/2Z, i.e., the
set of integers {0, 1} where addition and multiplication is
modulo 2.
The polynomial dynamical system for our model (cor-
responding to the update rules listed in Table 2) is: F =
( f1, . . . , f22) : F22
2
→F22
2
where the variables xi, i =
1, . . . , 22 are the species and the fi, i = 1, . . . , 22 are
the update functions written as the following polynomials
over F2.
x1 = hapX
x2 = sreA
x3 = HapX
x4 = SreA
x5 = RIA
x6 = EstB
x7 = MirB
x8 = SidA
x9 = TAFC
x10 = ICP
x11 = LIP
x12 = CccA
x13 = FC−Fe
x14 = FC+Fe
x15 = VAC
x16 = ROS
x17 = Yap1
x18 = SOD2/3
x19 = Cat1/2
x20 = ThP
x21 = Fe3+
x22 = O−
2
f1 = x4 + 1
f2 = x3 + 1
f3 = x11 · x1 + x1
f4 = x11 · x2
f5 = x4 + 1
f6 = x4 + 1
f7 = x3 · x4 + x3
f8 = x3 · x4 + x3
f9 = x8
f10 = x3 · x14 · x15 + x3 · x14 + x3 · x15 + x14 · x15
+ x14 + x15
f11 = x21 · x5 · x9 · x7 · x6 + x9 · x7 · x6 + x21 · x5
f12 = x3 + 1
f13 = x8
f14 = x11 · x13
f15 = x11 · x12
f16 = x16 · x11 · x22 · x18 · x19 · x20
+ x16 · x11 · x22 · x18 · x19 + x16 · x11 · x22 · x18 · x20
+ x16 · x11 · x18 · x19 · x20 + x16 · x22 · x18 · x19 · x20
+ x11 · x22 · x18 · x19 · x20 + x16 · x11 · x18 · x19
+ x16 · x22 · x18 · x19 + x16 · x11 · x18 · x20
+ x16 · x22 · x18 · x20 + x16 · x18 · x19 · x20
+ x22 · x18 · x19 · x20 + x16 · x11 · x22 + x16 · x18 · x19
+ x16 · x18 · x20 + x16 · x11 + x16 · x22 + x11 · x22
+ x16 + x11 + x22
f17 = x16
f18 = x17
f19 = x3 · x17 + x17
f20 = x17
f21 = x21
f22 = x22
Incorporating stochasticity
In this study, we use a random update schedule to simulate
dynamic behavior. The basic idea behind this approach is
to change the deterministic update of each species into
a probability of being updated. The stochastic discrete
dynamical systems (SDDS) framework was used to gen-
erate stochastic simulations [76]. An SDDS is a time- and
state-discrete dynamical system which models stochas-
ticity at the functional level by introducing two update
probabilities that, together with the update function, spec-
ify a probability of transition of a given species at each
time step. Let pin↑be the probability that species xi will
be updated given that the corresponding update func-
tion fi specifies an increase in state at the next time step.
Let p↓
i be the probability xi will be updated given that fi
specifies a decrease in state at the next time step. Then
a stochastic discrete dynamical system in n variables is a
collection of triplets
fi, p↑
i , p↓
i
n
i=1 where we may repre-
sent the update functions fi as polynomials over a finite
field. Thus a SDDS can be represented as a PDS along with
propensity parameters.
The probabilities p↑
i , p↓
i ∈[ 0, 1] for all i ∈{1, . . . , n} are
called the activation propensity and degradation propen-
sity, respectively, of the i-th species. If p↑
i = p↓
i = 1 for all
i = 1, . . . , n then all species are updated simultaneously
at every time step, and the simulation is deterministic. To
implement a random update schedule, we let p↑
i = p↓
i =
0.5 for all i = 1, .., n, meaning that at each time step each
species has an equal probability of either being updated
or remaining in its current state. Hence at each time
step, some species are randomly selected to be updated
whereas others are not. Updating of selected species is
done simultaneously at each time step. A “time step” in
this model refers to a single round of updates in which the
Brandon et al. BMC Systems Biology (2015) 9:19
Page 15 of 17
state of any given species can be updated only once. The
unit of time step is arbitrary, yet based on comparisons
with experimental time course data (Figure 4), we deter-
mined each time step of our model corresponds to about
6 minutes of real time.
Analysis of Dynamic Algebraic Models (ADAM), a free
web-based software tool which analyzes the dynamics of
discrete models using Gröbner bases calculations, was
used to generate the above PDS from transition tables and
to simulate dynamic behavior using the SDDS framework
[77]. ADAM is available at http://adam.plantsimlab.org/.
Experimental methods
A. fumigatus strain and growth conditions
The A. fumigatus strain used was wild-type AF293. A.
fumigatus was cultured on glucose minimal media plus
agar plates at 37°C for 7 to 10 days until fully conidiated.
Spores were harvested by flooding the culture plates with
endotoxin-free phosphate-buffered saline solution con-
taining 0.05% Tween-20 and swabbing with a sterile inoc-
ulation loop to obtain spore suspension. The spores were
vortexed and concentrations of spores were determined
by counting with a hemacytometer.
Incubation and harvesting
A. fumigatus was grown in a liquid shaker under iron
depleted conditions. 25 × 106 A. fumigatus condia were
added to standard glucose minimal media plus 0.05%
Tween-20 but without iron in the trace elements to a final
volume of a 25 mL for a final concentration of 1 million
spores per mL. Flasks were incubated at 37°C and 200
rpm for 72 hours. Glass flasks were rinsed prior to inoc-
ulation with a 0.1 M HCL solution followed by a rinse
with double distilled water to remove residual traces of
iron. After 72 hours, A. fumigatus was shifted from iron
depleted to iron replete conditions by adding FeSO4 to
a final concentration of 10μM FeSO4. A. fumigatus was
then incubated for another 9 hours.
Mycelia were harvested from triplicate samples at 0
(control), 30, 60, 90, 120, 150, 180, 210, 240, 270, 300, 330,
and 360 minutes after the addition of iron. Mycelia were
filtered through gauze and immediately flash frozen in
liquid nitrogen and stored at -80°C. Frozen mycelia were
subsequently ground to a fine powder using a mortar and
pestle in the presence of liquid nitrogen.
RNA extraction and cDNA synthesis
Total RNA was isolated using a Qiagen RNeasy plant
mini-kit. “Protocol: Purification of Total RNA from Plant
Cells and Tissues and Filamentous Fungi” was used along
with optional on-column DNase digestion step. Extracted
RNA was stored at -80°C. RNA integrity was assessed
by gel electrophoresis. Concentrations of RNA in each
sample were determined by spectrophotometry on a
Table 4 Primers used for real-time qRT-PCR
Gene
Primer sequence (5’-3’)
melting
Product
Tm (°C)
size(bp)
β-tubuliln
FP
CTGCTCTGCCATTTTCCGTG
56.8
119
RP
CGGTCTGGATGTTGTTGGGA
57.3
sidA
FP
TGACGACTCGCCTTTTGTGAA
57.0
474
RP
TTGCTCGGGTCCATCTCAAC
57.3
sreA
FP
CTCAGTACGATCGCTTCCCC
57.3
297
RP
GTCCCACAATTACTGCACGA
55.2
ftrA
FP
GGCATGATCGGAGCGTTCTA
57.1
411
RP
GGCTTGGTTTCCTCCTCGAT
57.2
cccA
FP
GAGCCAAGAGTGAGGCAGAA
57.0
448
RP
TGCACACCACCCTTGATACC
57.4
NANODROP 1000 Spectrophotometer. Next, cDNA was
synthesized following manufacturer’s instructions (Tetro
cDNA Synthesis Kit, Bioline). All incubations were car-
ried out in a thermacycler. Following synthesis, cDNA was
stored at -20°C.
qRT-PCR
Real time reverse transcription polymerase chain reaction
(qRT-PCR) was performed using the cDNA as a template.
The constitutively expressed gene β-tubulin of A. fumi-
gatus was used as the house-keeping gene. See Table 4
for a list of primers for target genes. Real time qRT-PCR
was carried out in 20 μL reaction volumes on a BIO-
RAD iQTM5 Multicolor Real-Time PCR Detection System
machine. The real time qRT-PCR consisted of the follow-
ing a 3-step protocol: (95°C denaturation for 10 s, 55°C
annealing period for 30 s, 72°C extension for 45 s) × 40
cycles. Cycling involved an initial denaturing/polymerase
activation step (95°C for 3 min) and a final melting curve
analysis (+0.5°C ramping × 81 cycles; 30 second incu-
bation between each cycle). SYBR Green (Bioline) was
used as the fluorescent reporter molecule in all reactions.
Real time qRT-PCR mixes consisted of 1 μL template
cDNA to 19 μL master mix. Relative gene expression (fold
change from the addition of iron) was quantified using the
Pfaffl method and normalized to β-tubulin [78]. Results
were collected from biological triplicates, and qRT-PCR
for each biological replicate was carried out in techni-
cal duplicates. Standard errors were calculated to ensure
statistical accuracy.
Additional file
Additional file 1: Brandon2015_Aspergillus_iron_superoxide. The
Boolean network model of Aspergillus fumigatus iron acquisition and
oxidative stress response is provided in SBML qual format.
Brandon et al. BMC Systems Biology (2015) 9:19
Page 16 of 17
Competing interests
The authors declare that they have no competing interests.
Authors’ contributions
Conceived the study: RL and CL. Performed construction and analysis of
mathematical model: MB. Designed the qRT-PCR experiment: CL, BH, and MB.
Performed wet lab work: MB, BH. Performed data analysis: MB. Interpreted
results: MB, CL, RL. Wrote the manuscript: MB. Reviewed and edited manuscript:
BH, CL, and RL. All authors have read and approved the final manuscript.
Acknowledgements
We would like to acknowledge Drs. R. Cramer and H. Haas for meaningful
discussions. This work was supported in part by NIH/NIAID grant number
1R21AI101619 to RL and CL.
Author details
1Center for Cell Analysis and Modeling, University of Connecticut Health
Center, 400 Farmington Ave, 06030 Farmington, USA. 2Center for Quantitative
Medicine, University of Connecticut Health Center, 195 Farmington Ave, 06030
Farmington, USA. 3Department of Biological Sciences, Virginia Tech, 1405
Perry Street, 24061 Blacksburg, USA. 4Virginia Bioinformatics Institute, Virginia
Tech, 1015 Life Science Circle, 24061 Blacksburg, US. 5The Jackson Laboratory
for Genomic Medicine, 10 Discovery Drive, 06030 Farmington, USA.
6Department of Cell Biology, University of Connecticut Health Center, 263
Farmington Ave, 06030 Farmington, USA.
Received: 15 December 2014 Accepted: 31 March 2015
References
1.
Latgé JP. Aspergillus fumigatus and aspergillosis. Clinical Microbiol Rev.
1999;12(2):310–50.
2.
Lass-Flörl C, Roilides E, Löffler J, Wilflingseder D, Romani L. Minireview:
host defence in invasive aspergillosis. Mycoses. 2013;56(4):403–13.
3.
Henriet S, Verweij PE, Holland SM, Warris A. Invasive fungal infections in
patients with chronic granulomatous disease. Adv Exp Med Biol.
2013;764:27–55.
4.
Lanternier F, Cypowyj S, Picard C, Bustamante J, Lortholary O, Casanova
J-L, et al. Primary immunodeficiencies underlying fungal infections.
Current Opin Pediatr. 2013;25(6):736–47.
5.
Hissen AH, Wan AN, Warwas ML, Pinto LJ, Moore MM. The aspergillus
fumigatus siderophore biosynthetic gene sida, encoding l-ornithine
n5-oxygenase, is required for virulence. Infect Immun. 2005;73(9):
5493–503.
6.
Lin SJ, Schranz J, Teutsch SM. Aspergillosis case-fatality rate: systematic
review of the literature. Clin Infect Dis. 2001;32(3):358–66.
7.
Thomas PA. Fungal infections of the cornea. Eye (London, England).
2003;17(8):852–62.
8.
Schrettl M, Bignell E, Kragl C, Joechl C, Rogers T, Haynes K, et al.
Siderophore biosynthesis but not reductive iron assimilation is essential
for aspergillus fumigatus virulence. J Exp Med. 2004;200(9):1213–9.
9.
Paris S, Wysong D, Debeaupuis J-P, Shibuya K, Philippe B, Diamond RD,
et al. Catalases of aspergillus fumigatus. Infect Immun. 2003;71(6):
3551–62.
10. Leal SM, Vareechon C, Cowden S, Cobb BA, Latgé J-P, Momany M, et al.
Fungal antioxidant pathways promote survival against neutrophils during
infection. J Clin Invest. 2012;122(7):2482–98.
11. Kontoyiannis DP, Chamilos G, Lewis RE, Giralt S, Cortes J, Raad II, et al.
Increased bone marrow iron stores is an independent risk factor for
invasive aspergillosis in patients with high-risk hematologic malignancies
and recipients of allogeneic hematopoietic stem cell transplantation.
Cancer. 2007;110(6):1303–6.
12. Leal SM, Roy S, Vareechon C, Carrion SD, Clark H, Lopez-Berges MS, et al.
Targeting iron acquisition blocks infection with the fungal pathogens
aspergillus fumigatus and fusarium oxysporum. PLoS Pathogens.
2013;9(7):1003436.
13. Schrettl M, Kim HS, Eisendle M, Kragl C, Nierman WC, Heinekamp T, et al.
Srea-mediated iron regulation in aspergillus fumigatus. Mol Microbiol.
2008;70(1):27–43.
14. Eisendle M, Schrettl M, Kragl C, Müller D, Illmer P, Haas H. The
intracellular siderophore ferricrocin is involved in iron storage,
oxidative-stress resistance, germination, and sexual development in
aspergillus nidulans. Eukaryot Cell. 2006;5(10):1596–603.
15. Blaiseau PL, Lesuisse E, Camadro JM. Aft2p, a novel iron-regulated
transcription activator that modulates, with aft1p, intracellular iron use
and resistance to oxidative stress in yeast. J Biol Chem. 2001;276(36):
34221–6.
16. Röhm M, Grimm MJ, D’Auria AC, Almyroudis NG, Segal BH, Urban CF.
Nadph oxidase promotes neutrophil extracellular trap formation in
pulmonary aspergillosis. Infect Immun. 2014;82(5):1766–77.
17. Philippe B, Ibrahim-Granet O, Prévost MC, Gougerot-Pocidalo MA,
Sanchez Perez M, Van der Meeren A, Latgé JP. Killing of aspergillus
fumigatus by alveolar macrophages is mediated by reactive oxidant
intermediates. Infect Immun. 2003;71(6):3034–42.
18. Bonnett CR, Cornish EJ, Harmsen AG, Burritt JB. Early neutrophil
recruitment and aggregation in the murine lung inhibit germination of
aspergillus fumigatus conidia. Infect Immun. 2006;74(12):6528–39.
19. Sugui JA, Kim HS, Zarember KA, Chang YC, Gallin JI, Nierman WC, et al.
Genes differentially expressed in conidia and hyphae of aspergillus
fumigatus upon exposure to human neutrophils. PloS One. 2008;3(7):
2655.
20. Lambou K, Lamarre C, Beau R, Dufour N, Latge J-P. Functional analysis
of the superoxide dismutase family in aspergillus fumigatus. Mol
Microbiol. 2010;75(4):910–23.
21. Amir A, Meshner S, Beatus T, Stavans J. Damped oscillations in the
adaptive response of the iron homeostasis network of e. coli. Mol
Microbiol. 2010;76(2):428–36.
22. Theurl I, Fritsche G, Ludwiczek S, Garimorth K, Bellmann-Weiler R,
Weiss G. The macrophage: a cellular factory at the interphase between
iron and immunity for the control of infections. Biometals. 2005;18(4):
359–67.
23. Zarember KA, Sugui JA, Chang YC, Kwon-Chung KJ, Gallin JI. Human
polymorphonuclear leukocytes inhibit aspergillus fumigatus conidial
growth by lactoferrin-mediated iron depletion. J Immunol (Baltimore,
Md.: 1950). 2007;178(10):6367–73.
24. Fluckinger M, Haas H, Merschak P, Glasgow BJ, Redl B. Human tear
lipocalin exhibits antimicrobial activity by scavenging microbial
siderophores. Antimicrobial Agents Chemother. 2004;48(9):
3367–72.
25. Skaar EP. The battle for iron between bacterial pathogens and their
vertebrate hosts. PLoS Pathogens. 2010;6(8):1000949.
26. Weinberg ED. Iron availability and infection. Biochimica et biophysica
acta. 2009;1790(7):600–5.
27. Haas H. Iron - a key nexus in the virulence of aspergillus fumigatus. Front
Microbiol. 2012;3:28.
28. Moore MM. The crucial role of iron uptake in aspergillus fumigatus
virulence. Curr Opin Microbiol. 2013;16(6):692–9.
29. Schrettl M, Beckmann N, Varga J, Heinekamp T, Jacobsen ID, Jöchl C,
et al. Hapx-mediated adaption to iron starvation is crucial for virulence of
aspergillus fumigatus. PLoS Pathog;6(9):e1001124.
30. Hsiang T, Baillie DL. Comparison of the yeast proteome to other
fungal genomes to find core fungal genes. J Mol Evol. 2005;60(4):
475–83.
31. Miller MJ, Zhu H, Xu Y, Wu C, Walz AJ, Vergne A, et al. Utilization of
microbial iron assimilation processes for the development of new
antibiotics and inspiration for the design of new anticancer agents.
Biometals. 2009;22(1):61–75.
32. Wencewicz TA, Long TE, Möllmann U, Miller MJ. Trihydroxamate
siderophore-fluoroquinolone conjugates are selective sideromycin
antibiotics that target staphylococcus aureus. Bioconjugate Chem.
2013;24(3):473–86.
33. Schrettl M, Bignell E, Kragl C, Sabiha Y, Loss O, Eisendle M, et al. Distinct
roles for intra- and extracellular siderophores during aspergillus
fumigatus infection. PLoS Pathog. 2007;3(9):1195–207.
34. Linde J, Hortschansky P, Fazius E, Brakhage AA, Guthke R, Haas H.
Regulatory interactions for iron homeostasis in aspergillus fumigatus
inferred by a systems biology approach. BMC Syst Biol. 2012;6:6.
35. Fréalle E, Aliouat-Denis C-M, Delhaes L, Hot D, Dei-Cas E. Transcriptomic
insights into the oxidative response of stress-exposed aspergillus
fumigatus. Current Pharm Des. 2013;19(20):3713–37.
Brandon et al. BMC Systems Biology (2015) 9:19
Page 17 of 17
36. Saez-Rodriguez J, Simeoni L, Lindquist JA, Hemenway R, Bommhardt U,
Arndt B, et al. A logical model provides insights into t cell receptor
signaling. PLoS Comput Biol. 2007;3(8):163.
37. Bornholdt S. Boolean network models of cellular regulation: prospects
and limitations. J R Soc Interface R Soc. 2008;5 Suppl 1:85–94.
38. Liang J, Han J. Stochastic boolean networks: an efficient approach to
modeling gene regulatory networks. BMC Syst Biol. 2012;6:113.
39. Berestovsky N, Zhou W, Nagrath D, Nakhleh L. Modeling integrated
cellular machinery using hybrid petri-boolean networks. PLoS Comput
Biol. 2013;9(11):1003306.
40. Miskov-Zivanov N, Turner MS, Kane LP, Morel PA, Faeder JR. The
duration of t cell stimulation is a critical determinant of cell fate and
plasticity. Sci Signaling. 2013;6(300):97.
41. Albert R, Thakar J. Boolean modeling: a logic-based dynamic approach
for understanding signaling and regulatory networks and for making
useful predictions. Wiley interdiscip Rev Syst Biol Med. 2014;6(5):
353–69.
42. Franke R, Müller M, Wundrack N, Gilles E-D, Klamt S, Kähne T, et al.
Host-pathogen systems biology: logical modelling of hepatocyte growth
factor and helicobacter pylori induced c-met signal transduction. BMC
Syst Biol. 2008;2:4.
43. Thakar J, Saadatpour-Moghaddam A, Harvill ET, Albert R.
Constraint-based network model of pathogen-immune system
interactions. J R Soc Interface/R Soc. 2009;6(36):599–612.
44. Schaadt NS, Steinbach A, Hartmann RW, Helms V. Rule-based regulatory
and metabolic model for quorum sensing in p. aeruginosa. BMC Syst Biol.
2013;7:81.
45. Bähler J, Svetina S. A logical circuit for the regulation of fission yeast
growth modes. J Theoretical Biol. 2005;237(2):210–18.
46. Davidich MI, Bornholdt S. Boolean network model predicts knockout
mutant phenotypes of fission yeast. PloS One. 2013;8(9):71786.
47. Awad S, Chen J. Inferring transcription factor collaborations in gene
regulatory networks. BMC Syst Biol. 2014;8 Suppl 1:1.
48. Achcar F, Camadro J-M, Mestivier D. A boolean probabilistic model of
metabolic adaptation to oxygen in relation to iron homeostasis and
oxidative stress. BMC Syst Biol. 2011;5:51.
49. Wallner A, Blatzer M, Schrettl M, Sarg B, Lindner H, Haas H. Ferricrocin, a
siderophore involved in intra- and transcellular iron distribution in
aspergillus fumigatus. Appl Environ Microbiol. 2009;75(12):4194–6.
50. Nilius AM, Farmer SG. Identification of extracellular siderophores of
pathogenic strains of aspergillus fumigatus. J Med Vet Mycol. 1990;28(5):
395–403.
51. Haas H, Eisendle M, Turgeon BG. Siderophores in fungal physiology and
virulence. Annu Rev Phytopathol. 2008;46:149–87.
52. Blatzer M, Binder U, Haas H. The metalloreductase freb is involved in
adaptation of aspergillus fumigatus to iron starvation. Fungal Genet Biol.
2011;48(11):1027–33.
53. Hissen AH, Chow JM, Pinto LJ, Moore MM. Survival of aspergillus
fumigatus in serum involves removal of iron from transferrin: the role of
siderophores. Infect Immun. 2004;72(3):1402–8.
54. Raymond-Bouchard I, Carroll CS, Nesbitt JR, Henry KA, Pinto LJ,
Moinzadeh M, et al. Structural requirements for the activity of the mirb
ferrisiderophore transporter of aspergillus fumigatus. Eukaryot Cell.
2012;11(11):1333–44.
55. Kragl C, Schrettl M, Abt B, Sarg B, Lindner HH, Haas H. Estb-mediated
hydrolysis of the siderophore triacetylfusarinine c optimizes iron uptake
of aspergillus fumigatus. Eukaryot Cell. 2007;6(8):1278–85.
56. Gsaller F, Eisendle M, Lechner BE, Schrettl M, Lindner H, Müller D, et al.
The interplay between vacuolar and siderophore-mediated iron storage
in aspergillus fumigatus. Metallomics. 2012;4(12):1262–70.
57. Eisendle M, Oberegger H, Zadra I, Haas H. The siderophore system is
essential for viability of aspergillus nidulans: functional analysis of two
genes encoding l-ornithine n 5-monooxygenase (sida) and a
non-ribosomal peptide synthetase (sidc). Mol Microbiol. 2003;49(2):
359–75.
58. Mercier A, Labbé S. Both php4 function and subcellular localization are
regulated by iron via a multistep mechanism involving the glutaredoxin
grx4 and the exportin crm1. J Biol Chem. 2009;284(30):20249–62.
59. Jbel M, Mercier A, Labbé S. Grx4 monothiol glutaredoxin is required for
iron limitation-dependent inhibition of fep1. Eukaryot Cell. 2011;10(5):
629–45.
60. Kim K-D, Kim H-J, Lee K-C, Roe J-H. Multi-domain cgfs-type glutaredoxin
grx4 regulates iron homeostasis via direct interaction with a repressor
fep1 in fission yeast. Biochem Biophys Res Commun. 2011;408(4):609–14.
61. Lessing F, Kniemeyer O, Wozniok I, Loeffler J, Kurzai O, Haertl A, et al.
The aspergillus fumigatus transcriptional regulator afyap1 represents the
major regulator for defense against reactive oxygen intermediates but is
dispensable for pathogenicity in an intranasal mouse infection model.
Eukaryot Cell. 2007;6(12):2290–302.
62. Qiao J, Kontoyiannis DP, Calderone R, Li D, Ma Y, Wan Z, et al. Afyap1,
encoding a bzip transcriptional factor of aspergillus fumigatus,
contributes to oxidative stress response but is not essential to the
virulence of this pathogen in mice immunosuppressed by
cyclophosphamide and triamcinolone. Med Mycol. 2008;46(8):773–82.
63. Dixon SJ, Stockwell BR. The role of iron and reactive oxygen species in
cell death. Nat Chem Biol. 2014;10(1):9–17.
64. Thön M, Al-Abdallah Q, Hortschansky P, Brakhage AA. The thioredoxin
system of the filamentous fungus aspergillus nidulans: impact on
development and oxidative stress response. J Biol Chem. 2007;282(37):
27259–69.
65. Chaouiya C, Bérenguier D, Keating SM, Naldi A, van Iersel MP,
Rodriguez N, et al. Sbml qualitative models: a model representation
format and infrastructure to foster interactions between qualitative
modelling formalisms and tools. BMC Syst Biol. 2013;7:135.
66. Elowitz MB, Levine AJ, Siggia ED, Swain PS. Stochastic gene expression
in a single cell. Science (New York, N.Y.) 2002;297(5584):1183–6.
67. Swain PS, Elowitz MB, Siggia ED. Intrinsic and extrinsic contributions to
stochasticity in gene expression. Proc Nat Acad Sci USA. 2002;99(20):
12795–800.
68. Ozbudak EM, Thattai M, Kurtser I, Grossman AD, van Oudenaarden A.
Regulation of noise in the expression of a single gene. Nat Genet.
2002;31(1):69–73.
69. Ko MS. Induction mechanism of a single gene molecule: stochastic or
deterministic? BioEssays. 1992;14(5):341–46.
70. Munsky B, Neuert G, van Oudenaarden A. Using gene expression noise
to understand gene regulation. Science (New York, N.Y.) 2012;336(6078):
183–7.
71. Singh A. Transient changes in intercellular protein variability identify
sources of noise in gene expression. Biophys J. 2014;107(9):2214–20.
72. Power T, Ortoneda M, Morrissey JP, Dobson AD. Differential expression
of genes involved in iron metabolism in aspergillus fumigatus. Int
Microbiol. 2006;9(4):281–7.
73. Perrin RM, Fedorova ND, Bok JW, Cramer RA, Wortman JR, Kim HS, et al.
Transcriptional regulation of chemical diversity in aspergillus fumigatus
by laea. PLoS Pathogens. 2007;3(4):50.
74. Laubenbacher R, Stigler B. A computational algebra approach to the
reverse engineering of gene regulatory networks. J Theor Biol.
2004;229(4):523–37.
75. Lidl R, Niederreiter H. Finite Fields. New York: Cambridge University Press;
1997.
76. Murrugarra D, Veliz-Cuba A, Aguilar B, Arat S, Laubenbacher R.
Modeling stochasticity and variability in gene regulatory networks.
EURASIP J Bioinform Syst Biol. 2012;2012(1):5.
77. Hinkelmann F, Brandon M, Guang B, McNeill R, Blekherman G,
Veliz-Cuba A, et al. Adam: analysis of discrete models of biological
systems using computer algebra. BMC Bioinformatics. 2011;12:295.
78. Pfaffl MW. A new mathematical model for relative quantification in
real-time rt-pcr. Nucleic Acids Res. 2001;29(9):45.
|
25908096
|
ThP = ( Yap1 )
Cat1/2 = ( ( Yap1 ) AND NOT ( HapX ) )
ROS = ( ( Superoxide ) AND NOT ( SOD2/3 AND ( ( ( ThP AND Cat1/2 ) ) ) ) ) OR ( LIP ) OR ( ( ROS ) AND NOT ( SOD2/3 AND ( ( ( ThP AND Cat1/2 ) ) ) ) )
ICP = ( ( VAC ) AND NOT ( HapX ) ) OR ( ( FCplusFe ) AND NOT ( HapX ) )
FCminusFe = ( SidA )
SreA = ( LIP AND ( ( ( sreA ) ) ) )
sreA = NOT ( ( HapX ) )
SidA = ( ( HapX ) AND NOT ( SreA ) )
TAFC = ( SidA )
HapX = ( ( hapX ) AND NOT ( LIP ) )
LIP = ( RIA AND ( ( ( Iron ) ) ) ) OR ( TAFC AND ( ( ( EstB AND MirB ) ) ) )
CccA = NOT ( ( HapX ) )
EstB = NOT ( ( SreA ) )
Yap1 = ( ROS )
hapX = NOT ( ( SreA ) )
VAC = ( CccA AND ( ( ( LIP ) ) ) )
MirB = ( ( HapX ) AND NOT ( SreA ) )
SOD2/3 = ( Yap1 )
RIA = NOT ( ( SreA ) )
FCplusFe = ( FCminusFe AND ( ( ( LIP ) ) ) )
|
RESEARCH
Open Access
Proteins interaction network and modeling
of IGVH mutational status in chronic
lymphocytic leukemia
María Camila Álvarez-Silva1†, Sally Yepes2†, Maria Mercedes Torres2 and Andrés Fernando González Barrios1*
* Correspondence:
andgonza@uniandes.edu.co
†Equal contributors
1Grupo de Diseño de Productos y
Procesos (GDPP), Departamento de
Ingeniería Química, Universidad de
los Andes, Bogotá, DC, Colombia
Full list of author information is
available at the end of the article
Abstract
Background: Chronic lymphocytic leukemia (CLL) is an incurable malignancy of
mature B-lymphocytes, characterized as being a heterogeneous disease with variable
clinical manifestation and survival. Mutational statuses of rearranged immunoglobulin
heavy chain variable (IGVH) genes has been consider one of the most important
prognostic factors in CLL, but despite of its proven value to predict the course of the
disease, the regulatory programs and biological mechanisms responsible for the
differences in clinical behavior are poorly understood.
Methods: In this study, (i) we performed differential gene expression analysis
between the IGVH statuses using multiple and independent CLL cohorts in
microarrays platforms, based on this information, (ii) we constructed a simplified
protein-protein interaction (PPI) network and (iii) investigated its structure and
critical genes. This provided the basis to (iv) develop a Boolean model, (v) infer
biological regulatory mechanism and (vi) performed perturbation simulations in
order to analyze the network in dynamic state.
Results: The result of topological analysis and the Boolean model showed that the
transcriptional relationships of IGVH mutational status were determined by specific
regulatory proteins (PTEN, FOS, EGR1, TNF, TGFBR3, IFGR2 and LPL). The dynamics
of the network was controlled by attractors whose genes were involved in
multiple and diverse signaling pathways, which may suggest a variety of
mechanisms related with progression occurring over time in the disease. The
overexpression of FOS and TNF fixed the fate of the system as they can activate
important genes implicated in the regulation of process of adhesion, apoptosis,
immune response, cell proliferation and other signaling pathways related with
cancer.
Conclusion: The differences in prognosis prediction of the IGVH mutational status
are related with several regulatory hubs that determine the dynamic of the system.
Keywords: CLL, PPI network, Boolean network, Topological analysis
Background
Chronic lymphocytic leukemia (CLL), the most common type of adult leukemia in de-
veloped countries, is an incurable malignancy of mature B lymphocytes, characterized
by accumulation of mature B cells in the blood, bone marrow, and secondary lymphoid
organs such as the lymph nodes (LN) [1, 2]. Patients with CLL show a highly variable
© 2015 Alvarez-Silva et al. This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided
the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/
publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
DOI 10.1186/s12976-015-0008-z
disease evolution and different response to therapy. This variability may be related to
evolutionary dynamics of sub-clonal mutations [3]. Investigations of the B cell receptor
(BCR) indicate that 60–65 % of CLLs carry immunoglobulin heavy chain variable (IGHV)
genes with evidence of somatic hypermutation and this may modify BCR affinity for
antigens. Conversely, 35–40 % of CLLs are devoid of IGHV somatic mutations [4].
Understanding the pathological mechanisms of CLL has helped to divide the disease
into two risk categories that have a strong impact on prognosis and treatment: 1)
patients with minimal clinical manifestations and 2) an aggressive form characterized
by high mortality, whose IGHV genes can be somatically mutated or unmutated,
respectively. Due to the importance of IGVH status in the determination of the course
of the disease, several expression studies have focused on the comparison of CLL type
mutated IGVH vs. IGVH unmutated. Nevertheless, these studies have identified genes
that are not functionally related and therefore cannot elucidate biological mechanisms
to distinguish between risk categories.
The interactions of proteins are essential to execute biological functions in different
contexts [5]. Since cancer is a complex and multi-factorial disease involving diverse
anomalies, the representation and analysis of a malignant cell as a protein-protein
interaction (PPI) network can provide insights into its behavior. It has been postulated
that proteins with high connectivity within a PPI network could represent meaningful
biological information, despite non-being differentially expressed [6]. Thus, the inte-
grated analysis of gene expression data with PPI networks could be valuable method to
provide knowledge into molecular mechanisms of diseases. The analyses of PPI
networks have varied applications such as identification of drug targets, functional
protein modules and disease candidate genes [7, 8].
On the other hand, dynamic network modeling can be used to gain insight into the
functionality of biological processed and made possible simulations to predict models
behavior [9]. Modeling of regulatory networks as dynamical systems includes modeling
based on ordinary differential equations [10, 11], Bayesian framework [12] and Boolean
rules [13, 14]. Given the limitation of quantitative models that need knowledge on the
kinetics and mechanistic parameters of the system, in addition of a wealth of qualitative
and interaction data obtained from the experimental literature and high-throughput
technologies, the qualitative approaches such as Boolean modeling become an ex-
tremely useful resource [15].
The Boolean modeling considers the genes as binary variables being either active or
passive, but encompassing the essential functionality of the system, the general building
blocks that have been identified in Boolean networks constitute different types of
robust switching elements [16]. This type of approach is already successfully applied in
complex models as the FA/BRCA pathway in Fanconi anemia [17], the survival process
of in large granular lymphocyte leukemia [18], process of T-helper lymphocytes [19]
and the control of the mammalian cell cycle [20].
In this study, we constructed a simplified PPI network of the IGVH mutational status
in CLL and analyzed its structure and critical genes. We used the topology of the
network to develop a Boolean model, infer regulatory mechanisms and perform
simulations to analyze the network in dynamic state. The modeling of the PPI network
led to identify regulatory elements of the disease, contributing to understand the prog-
nostic differences and the dynamic behavior under different perturbations over time.
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 2 of 15
Results
We inquired into the impact of differentially expressed genes (DEG) between the IGVH
statuses by mapping them onto the PPI network. The initial data set of 502 DE genes
was reduced to 90 genes with the software STRING and exhaustive literature
reviewed. The PPI network reconstructed contains 90 nodes and 120 regulatory edges
(Fig. 1). An additional table shows the functions of all genes implicated in the network
(Additional file 1: Table S1). The resulting simplified network made evident that DEG
between IGVH statuses are not always represented by highly connected nodes.
The main topological characteristic of the network is the degree distribution P(k), which
tells us the probability that a select node has exactly k links. The degree distribution P(k)
allows us to distinguish between different types of biological networks. We obtained a
power-law degree distribution which characterize scale free networks (Fig. 2), where the
probability that a node displays k links follows P(k) ∼k−γ, where γ is the degree exponent
that describes the role of the hubs in the system [21]. In the PPI network we obtained
values of γ between 2 and 3, indicating that there exist a hierarchy of hubs, i.e. there are a
large number of nodes with few connections while highly connected nodes are scarce
[21]. The following genes showed the highest values in degree evaluation: in-degree
(PTEN, FOS, and EGR1) and out-degree (TNF, TGFBR3, and IFGR2). Values of γ between
2 and 3 refer also to the small-world property [22], characterized by a small value of diam-
eter, which increase the network efficiency. However, we obtained a value of diameter of
8, greater-than-expected for networks with the small-world property. To explain the
values of diameter higher to the expected, Zhang et al. [22] made a comparison between
the values of diameter reported for real biological networks regarding to diameters ob-
tained for networks with the same features in which the connections between nodes are
randomized. Specifically, they evaluated the diameter of protein-protein interaction
Fig. 1 Protein-protein interaction network. Pink edges: inhibition. Green edges: activation. Purple nodes: low
expression. Blue nodes: high expression
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 3 of 15
networks of biological organisms in contrast to the obtained in randomized networks.
Their simulation showed that an increment of the diameter in the real networks allows a
significant increase of the network modularity, suggesting an adjustment between network
efficiency and the benefits obtainable by modularity [22].
This analysis was completed by other centralities measures such as closeness and be-
tweenness, which provide a characterization of nodes that are relevant for the network
structure. Closeness is defined by the inverse of the average length of the shortest paths
to all other nodes [23]. We found TNF, TGFBR3, and IFGR2 as proteins with the high-
est closeness values in the PPI network. A protein with high closeness, compared to
the average closeness of the network, will be easily central to the regulation of other
proteins but with some proteins not influenced by its activity [24]. Implying that those
proteins are the closest to all other nodes and have an extent of influences on the entire
Number of nodes
Number of nodes
In-degree
Out-degree
a
b
Fig. 2 a. Power-law in-degree distribution b. Power-law out-degree distribution
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 4 of 15
network. This parameter can also be considered a measure of how long it will take in-
formation to spread from a given node to others [25]. Betweenness is defined by the
number of shortest paths that pass through a node [26]. The betweenness index favors
nodes that join communities rather than nodes that lie inside a community [27, 28].
FOS, TNF and LPL exhibited the highest values, implying a role as linkers in the
control of interactions between proteins. Selected genes, reported above, can be seen
in Fig. 3. The overall parameters that characterized the network are shown in the
Additional file 2: Table S2, and the topological parameters for each one of the nodes
are shown in the Additional file 3: Table S3.
We saw that centrality measures in the PPI network shown some degrees of overlap,
stressing the importance of the implicated proteins in the system structure. According
to Yu et al. [29], as a complementary notion of highly connected proteins known as
hubs proteins, it is possible to define bottlenecks proteins as proteins with high be-
tweenness values, bottlenecks proteins are essential connectors with surprising func-
tional and dynamic properties. Therefore, to develop a Boolean model and evaluate the
genes with influence in the behavior of the network over time, we focused on those that
were at the center of the major structural hubs and simultaneously exhibited the highest
values in the topological centralities evaluated. The selected nodes were: FOS, PTEN,
TGFBR3, and TNF. The dependencies between the genes obtained from the literature re-
view were translated to rule sets. The biological events for activation or inhibition were
qualitatively represented by Boolean functions, that is, combinations of AND, OR, and
NOT operations, that determine the evolution of a node through time and their relation
to the other components of the system (Additional file 4: Table S4).
Starting from an initial condition, the Boolean model evolved over time to finally
stabilizes in a recurrent state known as attractor, representing the long-term behavior
of the system [15]. We found a simple attractor for the PPI network for CLL consisting
of one state. The model obtained achieves the fixed point (steady state) after six time
steps. The state of transition and its successor attractor (starting from the initial state
determined by microarray analysis) are shown in Fig. 4. Starting from 50 random initial
states, the system had both the single-state attractors and cycle attractors. The major
cycle attractors displayed four states (Fig. 5). This showed important dependency of the
achieved attractor according to the initial system state.
Given the relevance of signaling pathways as triggers of processes associated with
cancer oncogenesis and progression, the activated proteins in the attractor were
subjected to modular functional enrichment to determine annotations from KEGG and
Panther
pathways
simultaneously.
The
associated
annotations
involved
various
pathways related to cancer with statistical significance for focal adhesion (corrected p
value = 0.000231747). Other signaling pathways detected in the attractor included: B
cell receptor, T cell receptor, cytokine-cytokine, integrin, and cadherin, among others.
Processes related with cell proliferation and regulations of transcription were also
present.
Given the overriding importance attributed to FOS and TNF in cancer biology and
their high topological measures in the PPI network, we chose these genes to performed
perturbation simulations of knockout and overexpression in the system. Furthermore,
we analyzed the effect of different initial conditions that can lead the system to different
steady states (attractors). The two initial conditions that were taken into account were: i)
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 5 of 15
initial condition determined by microarray analysis and ii) an initial state in which all
nodes were active.
The overexpression of FOS produced activation of genes related with multiples sig-
naling pathways. When comparing activated genes under the FOS overexpression re-
gardless of activated genes in the original attractor, the modular functional analysis
found the MAPK signaling pathway with the most significant value, (p = 4.37211e-05).
Other pathways involved were: apoptosis (p = 7.34148e-05), PDGF (p = 0.000100226),
JAK-STAT (p = 0.0001216), angiogenesis (p = 0.0001216) and cytokine-cytokine inter-
action (p = 0.000500178). Similarly, activated genes under TNF overexpression were
involved in multiple signaling pathways associated with cancer: focal adhesion (p =
7.05488e-07), angiogenesis (p = 8.39556e-07), JAK-STAT (p = 1.39118e-06), tight junc-
tion (p = 5.76719e-06), MAPK (p = 7.60619e-06), Wnt (p = 0.000105245), among others.
Under both initial conditions the effect of knocking out of any gene did not display
an effect, since the same attractor was obtained when no gene was knocked out. Never-
theless, the path length to achieve the attractor varies significantly, that is, depending
on the initial conditions, the system takes more or less steps of evolution in time to
stabilize in a recurrent state, known as attractor. From this behavior of the system, it
can be concluded that the attractor achieved is the most stable state of the network,
because although there are perturbations involving the system, the network returns to
the same steady state after different steps of time evolution.
On the other hand, when constantly maintaining the major nodes active under the
two initial conditions studied, we found that the selected gene displayed an effect on
the attractor achieved and therefore on the behavior of the system, affecting evolution
Percentage
Percentage
Percentage
Percentage
FOS
TNF
PTEN
TGFBR3
OutDegree InDegree Closeness Betweenness
centrality value average min max
OutDegree InDegree Closeness Betweenness
OutDegree InDegree Closeness Betweenness
OutDegree InDegree Closeness Betweenness
Fig. 3 Measure of centralities of FOS, TNF, PTEN and TGFBR3
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 6 of 15
of the network. These changes in the behavior are important to determine its relation
to the evolution of the disease.
Under both conditions of perturbation, the overexpression of the gene FOS and TNF
showed important influence in the evolution over time of the system (Fig. 6). In both
cases the system reaches complex attractors in which the network oscillates among a
set of four states, i.e., the attractor of the network is a cycle. In these states the nodes
involved in regulation of CLL oscillate under states of OFF or ON, which affects the
course of the disease.
Discussion
We applied a system approach by linking proteins interaction data with differentially
expressed genes of the IGVH status, the reconstructed PPI network allowed identified
critical genes, and given the stringent parameters applied, they represent only relation-
ships based on strong protein interactions. The topology of the reconstructed PPI net-
work followed the power-law of node degree distribution, a feature of true complex
biological networks. Therefore, the obtained network is a scale-free biological entity ra-
ther than a random network, indicating the presence of few nodes having a very high
degree measure [21]. On the other hand, it is recognized that high values of degree cen-
trality are associated with proteins that are interacting with several others suggesting a
central regulatory role [23]. The degree index highlighted some important proteins with
regulatory functions and considered interactions hubs in the PPI network: PTEN, FOS,
EGR1, TNF, TGFBR3, and IFGR2. According to the lethality and centrality rule, the
highly connected nodes are biologically relevant, representing vulnerable and essential
Inactive
Active
a
b
t= 1
2
3
4
5
6
steady state
Fig. 4 Visualization of a sequence of states. a. The columns of the table represent consecutive states of the
time series. b. Steady-state attractor of the network from initial state determined by microarray analysis. Genes
are encoded in the following order: AEBP1 AFF1 AICDA AKAP12 AKT3 ALOX5 ANXA2 APLP2 APOBEC3G APP
BLNK BMI1 CASP3 CAV1 CCL5 CCND2 CD27 CD63 CD69 CD70 CD79A CD81 CD86 CHST2 CNR1 CREM CSDA
CSNK2A2 CTSB CUL5 DPP4 EED EGR1 EZH2 FCER2 FGFR1 FOS FRK FYN GSK3B H2AFX HDAC9 HIST1H3H
HIST2H2AA3 HSP90AA1 HSP90B1 IFNGR2 IGF1R IL10RA IL7 ILK INPP5D JAK1 LGALS1 LIG1 LMNA LPL MAP2K6
MAP4K4 MARCKS MGAT5 MIF MYL9 MYLK NAB1 NCOR2 NFE2L2 NOTCH2 OGT PAX3 PCNA PLD1 PRF1 PRKCA
PTCH1 PTEN RFC5 RPS6KA5 RRM1 RUNX3 SELL SELP SIAH1 SKI TCF3 TNF TNFRSF1B VDR CD74 ADM TGFBR3.
Active genes in this attractor state were: AFF1, APLP2, APP, BMI1, CD27, CD81, CD86, CREM, CUL5, EED, EZH2,
FYN, GSK3B, HIST2H2AA3, HSP90B1, IL10RA, ILK, MARCKS, MGAT5, RRM1, SKI
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 7 of 15
points to system viability [30, 31], supporting this argument the establishment and
stability of cancer cells through these hubs.
It was noted that highly connected proteins in the PPI network are not necessarily
represented for highly DEG. Consequently, genes with a central role in cancer not
detected for high-throughput approaches could be identified by networks based analysis
[6]. This was the case for PTEN, FOS, EGR1 and TNF, whose p values were significant
but they were not among the lowest in the meta-analysis. Several genes with known
prognostic implications in CLL were present in the list of DEG; additionally, the top
DEG obtained were consistent with other studies [32–34], validating these findings the
microarray meta-analysis approach to produce robust conclusions.
LPL is one for the strongest prognostic markers to predict outcome in CLL [35, 36].
It was one of the genes with high betweenness index, reflecting the large amount of
control exerted by this node over the interactions between the other nodes, in this way,
LPL may function as bridge between sub-graphs. According to Kolset and Salmivirta
[37] LPL facilitate the contact between monocytes and endothelial cell through its
union with heparan sulfate proteoglycans, serving as a bridging protein between cell
surface proteins and lipoproteins. On the other hand, LPL may influence CLL behavior
for its relation with functional pathways involved in fatty acid degradation and signaling
[38].
All genes found with high centralities have central roles in cancer and are involved in
major, diverse and sometimes interrelated signaling pathways. They have roles as tumor
suppressors or oncogenes, engaging central role in cancer progression. PTEN has
phosphatase catalytic function that antagonizes the PI3K/AKT signaling pathway and
Inactive
Active
2.99% 2.45% 2.99% 2.72% 2.72% 2.99%
Fig. 5 Major attractors obtained from 50 random initial states. The columns of the table represent
consecutive states of the attractor. On top, the percentage of states leading to the attractor is supplied.
Genes are encoded in the following order: AEBP1 AFF1 AICDA AKAP12 AKT3 ALOX5 ANXA2 APLP2
APOBEC3G APP BLNK BMI1 CASP3 CAV1 CCL5 CCND2 CD27 CD63 CD69 CD70 CD79A CD81 CD86 CHST2
CNR1 CREM CSDA CSNK2A2 CTSB CUL5 DPP4 EED EGR1 EZH2 FCER2 FGFR1 FOS FRK FYN GSK3B H2AFX
HDAC9 HIST1H3H HIST2H2AA3 HSP90AA1 HSP90B1 IFNGR2 IGF1R IL10RA IL7 ILK INPP5D JAK1 LGALS1 LIG1
LMNA LPL MAP2K6 MAP4K4 MARCKS MGAT5 MIF MYL9 MYLK NAB1 NCOR2 NFE2L2 NOTCH2 OGT PAX3
PCNA PLD1 PRF1 PRKCA PTCH1 PTEN RFC5 RPS6KA5 RRM1 RUNX3 SELL SELP SIAH1 SKI TCF3 TNF TNFRSF1B
VDR CD74 ADM TGFBR3
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 8 of 15
suppresses cell survival as well as cell proliferation [39]. TNF gene encodes a multifunc-
tional proinflammatory cytokine that belongs to the tumor necrosis factor (TNF) super-
family, can induce a wide range of intracellular signal pathways including apoptosis and
cell survival as well as inflammation and immunity. TNF has two receptors (TNFR1,
TNFR2), TNFR1 signaling induces activation of many genes, primarily controlled by
two distinct pathways, NF-kappa B pathway and the MAPK cascade, or apoptosis and
necrosis. TNFR2 signaling activates NF-kappa B pathway, including PI3K-dependent
NF-kappa B pathway and JNK pathway leading to survival [40]. FOS gene family
encodes leucine zipper proteins that can dimerize with proteins of the JUN family,
thereby forming the transcription factor complex AP-1. As such, the FOS proteins have
been implicated as regulators of cell proliferation, differentiation, and transformation.
In some cases, expression of the FOS gene has also been associated with apoptotic cell
a
b
t=
1
2
3
4
t=
1
2
3
4
Inactive
Active
Fig. 6 Genes are encoded in the following order: AEBP1 AFF1 AICDA AKAP12 AKT3 ALOX5 ANXA2 APLP2
APOBEC3G APP BLNK BMI1 CASP3 CAV1 CCL5 CCND2 CD27 CD63 CD69 CD70 CD79A CD81 CD86 CHST2
CNR1 CREM CSDA CSNK2A2 CTSB CUL5 DPP4 EED EGR1 EZH2 FCER2 FGFR1 FOS FRK FYN GSK3B H2AFX
HDAC9 HIST1H3H HIST2H2AA3 HSP90AA1 HSP90B1 IFNGR2 IGF1R IL10RA IL7 ILK INPP5D JAK1 LGALS1 LIG1
LMNA LPL MAP2K6 MAP4K4 MARCKS MGAT5 MIF MYL9 MYLK NAB1 NCOR2 NFE2L2 NOTCH2 OGT PAX3
PCNA PLD1 PRF1 PRKCA PTCH1 PTEN RFC5 RPS6KA5 RRM1 RUNX3 SELL SELP SIAH1 SKI TCF3 TNF TNFRSF1B
VDR CD74 ADM TGFBR3. a. Visualization of states under overexpression of FOS b. Visualization of states
under overexpression of TNF
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 9 of 15
death [41]. Studies about EGR1 have been suggested that it is a cancer suppressor gene
and a transcriptional regulator. The products of target genes it activates are required
for differentiation and mitogenesis [42]. TGFBR3 is a membrane proteoglycan that
functions as a co-receptor with other transforming growth factors receptors. Soluble
TGFBR3 may inhibit TGFB signaling. Decreased expression of this receptor has been
observed in various cancers [41].
When the attractors found in the analysis of Boolean model are analyzed, several key
proteins associated with specific signaling pathways related with cancer are found, it
became clear that the phenotype depends upon multiple and interrelated signaling
pathways. We underscore the importance of MAPK signaling pathway, identified by
enrichment analysis, under FOS overexpression in the Boolean model. Belonging to the
MAPK/ERK signaling cascade were found activated: CASP3, FGFR1, AKT3, FOS,
MAP2K6, MAP4K4, PRKCA, RPS6KA5, and TNF. Aberrations in the MAPK/ERK
pathway
have
been
identified
in
human
cancers
in
high
frequency
including
hematologic malignancies [42]. In the context of CLL, the MAPK signaling pathway
has been recently implied in the disease based on clustering of RNA sequencing data
[43]. Similarly, working with gene co-expression subnetworks associated with disease
progression, it has been proven association of MAPK pathway with higher expression
levels in patients at early stages of the disease [44].
The regulatory hubs determine the behavior of the disease over time. The dynamics
of the network is controlled by attractors involved of diverse signaling pathways, which
may suggest a variety of mechanisms controlling the difference in CLL behavior. “The
2 distinct disease” hypothesis in CLL could be challenged; it is an interesting approach
to speculate that the CLL disease transcriptome evolves over time to reach a state
associated with disease requiring treatment [44]. These results have implications for
understanding transcriptional dynamic in the evolution of the disease.
Conclusion
The PPI network and a Boolean model of IGVH mutational status in CLL allowed iden-
tified regulatory proteins and generated insight about processes associated with the mani-
fest differences in prognosis. The perturbation in the network through overexpression of
important regulatory proteins, such as FOS and TNF, determine the dynamic of the
network and activate genes involved in different signaling pathways that play important
roles in cancer.
Methods
Differential expression analysis between the IGVH mutational statuses
To ensure reliability and generalization of results, we combined information from
different and independent microarray expression cohorts. It is well known that integra-
tion of expression data allow the discovery of new biological insights by increasing the
statistical power [45]. We retrieved CLL cohorts from the Gene Expression Omnibus
(GEO) of the National Center for Biotechnology Information (NCBI). The cohorts
selected (GSE2466, GSE16746, GSE9992 and GSE38611) had raw data available, were
originally processed with different microarray platforms and had at least 60 CLL
patients with information about the IGVH statuses, in total were processed 356 CLL
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 10 of 15
patients (174 mutated/umutated status). Each study was normalized independently
using the VSN method implemented in R [46]. For filtering non-expressed and non-
informative genes, matching genes among different microarray platforms and merging
among studies, we used MetaDE package in R [47]. To combining the information of
cohorts and avoid batch effect we followed a meta-analysis approach, the moderated-t
statstics with permutations was used for individual analysis and Fisher P-value combin-
ation method for combine the individual p values [48]. For meta-analysis we used the
“MetaOmics” software suite [47].
Reconstruction of protein–protein interaction network
The reconstruction of the protein–protein interaction network was based on data from
differential expression analysis between IGVH mutational statuses. The list of 502 DE
genes obtained was used to retrieve interacting partners from curated databases and
current literature.
The protein–protein interaction network was constructed based on the current litera-
ture and, through the STRING—Search Tool for the Retrieval of Interacting Genes/
Proteins—web source [49]. The STRING database contains information from several
sources, including experimental data, computational prediction methods, public text
collection and an recompilation of predicted protein interactions of databases such as
EXPASY [50], BIND [51], BioGRID [52], DIP [53], IntAct MINT [54], and HPRD [55]
and with interactions from the pathway databases such as PID [56], Reactome [57],
KEGG [58], and EcoCyc [59].
In order to reduce the amount of data while maintaining the main gene relations, the
parameters of confidence for STRING were restricted for obtain more reliable associa-
tions. Furthermore, the active prediction methods taken into account for STRING pre-
dictions were: co-expression, experiments, databases, and text mining. From the
protein-protein interactions predicted by STRING, with the restrictions mentioned
above, only the genetic relationships causing the activation or inhibition of the compo-
nents of the network were considered. In this way we reduced the number of network
nodes from 502 to 90. Is important to state that the database predictive methods could
reduce the overall confidence of network.
Network topology analysis
Topological analysis of the protein–protein interaction network was carried out by
plugin Network Analysis [60] of the open source program Cytoscape 3.1.0 [61].
A topological evaluation of the network was carried out by evaluating structural
parameters such as the clustering coefficient and degree distributions, to evaluate the
number of interactions among one node and its neighbors, normalized by the
maximum number of possible interactions, and to determine the number of nodes
directly connected (first neighbors) to a given node v, respectively. In the network
evaluation we distinguished in-degree distribution, when the edges target the node v,
and out-degree distribution, when the edges target the adjacent neighbors of v [62, 60].
The evaluation of the relevance of a protein in the PPI network was also made
through measurements of network centrality parameters, for this purpose, were used
the betweenness, closeness and degree metrics. On the other hand, to analyze the
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 11 of 15
“compactness” of the network, we evaluate the average path length and the network
diameter, parameters that indicate how distant are the two most distant nodes, showing
the overall proximity between nodes in the interaction network analyzed [21].
Boolean network model
In a Boolean network model, each node i = 1, 2, …, N represents a protein of the net-
work that can assume only binary states θi. When θi = 1 the protein is functionally ac-
tive (TRUE), on the other hand, when θi = 0 the protein is functionally inactive
(FALSE). Thus, a network with N nodes will have 2 N possible states [21]. In this
model, edges represent regulatory relationships between elements; their orientation in the
network follows the direction of regulation process from the upstream to the downstream
node. As time passes, the state of each node is determined by the states of its neighbor,
through Boolean transfer functions based on evidence from the literature [63, 64].
The exponential behavior of the possible states in a Boolean network makes it com-
putationally unsuitable for large networks, where it is necessary to reduce the network
size, restricting the protein–protein interactions to the relationships of activation or
inhibition of any of the components of the system.
In this study, the Boolean synchronous algorithm proposed by Stuart Alan Kauffman
in 1969 [13] was implemented. The synchronous pattern is the most simple update
mode, where the states of all nodes are updated simultaneously according to the last
state of the network [15]. We used the package BoolNet [65] to construct Boolean
synchronous networks from knowledge of the dependencies of genes, based on
evidence from the current literature.
Starting from an initial condition, the model evolves over time to finally stabilize in a
recurrent state known as attractor, representing the long-term behavior of the system
[15]. Different initial conditions for the model may lead the system to different attrac-
tors, whereby the Boolean model should start from previously known biological infor-
mation; in this model the initial states of the network were determined from results
supplied for microarray analysis of up and down regulation states (Additional file 4:
Table S4). Aiming to prove the sensitivity of the Boolean model, we assayed 50 random
initial states, and demonstrated that the constructed model shows changes in the struc-
ture of the attractor achieved in the steady state under different initial states.
To determine critical proteins for structure and behavior of the system, we examined
the changes in network attractors if a certain component is knocked out (fixed in the
OFF state in the Boolean model) or overexpressed (fixed in the ON state in the Boolean
model). If knocking out or overexpressing a component leads to changes in network
dynamic response, it can be concluded that this component is implicated in the
biological regulation processes [15].
Functional enrichment analysis
Significant concurrent annotations from KEGG and Panther pathways were searched
with GeneCodis software [66–68].
Additional files
Additional file 1: Table S1. Summary of genes functions.
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 12 of 15
Additional file 2: Table S2. Topological properties of the CLL network.
Additional file 3: Table S3. Topological parameters for the individual nodes.
Additional file 4: Table S4. Boolean transfer functions and initial conditions of the Boolean network.
Abbreviations
CLL: Chronic lymphocytic leukemia; IGVH: Immunoglobulin heavy chain variable; PPI: Protein-protein interaction;
DEG: Differentially expressed genes.
Competing interests
The authors declare that they have no competing interests.
Authors’ contributions
MCAS performed the Boolean model, topological analysis of the network, drafted the manuscript and analyzed data.
SY participated in the conception and design of the study, performed statistical analysis of microarrays, analyzed data,
and drafted the manuscript. AFGB participated in the design, coordination of the study, drafted the manuscript and
analyzed data. MMTC participated in the design and coordination of the study. All authors read and approved the final
manuscript.
Acknowledgements
Vicerectoria de Investigaciones and Facultad de Ciencias, Universidad de los Andes, supported this work.
Author details
1Grupo de Diseño de Productos y Procesos (GDPP), Departamento de Ingeniería Química, Universidad de los Andes,
Bogotá, DC, Colombia. 2Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá,
DC, Colombia.
Received: 12 March 2015 Accepted: 8 June 2015
References
1.
Murray F, Insel PA. Targeting cAMP in chronic lymphocytic leukemia: a pathway-dependent approach for the
treatment of leukemia and lymphoma. Expert Opin Ther Targets. 2013;17(8):937–49.
2.
Gaidano G, Foà R, Dalla-Favera R. Molecular pathogenesis of chronic lymphocytic leukemia. J Clin Invest.
2012;122(10):3432–8.
3.
Landau DA, Carter SL, Stojanov P, McKenna A, Stevenson K, Lawrence MS, et al. Evolution and impact of subclonal
mutations in chronic lymphocytic leukemia. Cell. 2013;152(4):714–26.
4.
Chiorazzi N, Ferrarini M. Cellular origin(s) of chronic lymphocytic leukemia: cautionary notes and additional
considerations and possibilities. Blood. 2011;117(6):1781–91.
5.
Wu J, Vallenius T, Ovaska K, Westermarck J, Mäkelä TP, Hautaniemi S. Integrated network analysis platform for
protein-protein interactions. Nat Methods. 2009;6(1):75–7.
6.
Sanz-Pamplona R, Berenguer A, Sole X, Cordero D, Crous-Bou M, Serra-Musach J, et al. Tools for protein-protein
interaction network analysis in cancer research. Clin Transl Oncol. 2012;14(1):3–14.
7.
Li Y, Li J. Disease gene identification by random walk on multigraphs merging heterogeneous genomic and
phenotype data. BMC Genomics. 2012;13 Suppl 7:S27.
8.
Sharan R, Ulitsky I, Shamir R. Network-based prediction of protein function. Mol Syst Biol. 2007;3:88. Epub 2007
Mar 13.
9.
Feiglin A, Hacohen A, Sarusi A, Fisher J, Unger R, Ofran Y. Static network structure can be used to model the
phenotypic effects of perturbations in regulatory networks. Bioinformatics. 2012;28(21):2811–8.
10.
Chen WW, Schoeberl B, Jasper PJ, Niepel M, Nielsen UB, Lauffenburger DA, et al. Input–output behavior of ErbB
signaling pathways as revealed by a mass action model trained against dynamic data. Mol Syst Biol. 2009;5:239.
11.
Jones RB, Gordus A, Krall JA, MacBeath G. A quantitative protein interaction network for the ErbB receptors using
protein microarrays. Nature. 2006;439(7073):168–74. Epub 2005 Nov 6.
12.
Grzegorczyk M, Husmeier D, Rahnenführer J. Modelling non-stationary dynamic gene regulatory processes with
the BGM model. Comput Stat. 2011;26:199–218.
13.
Kauffman SA. Metabolic stability and epigenesis in randomly constructed genetic nets. J Theor Biol.
1969;22(3):437–67.
14.
Kauffman SA. The origins of order: self-organization and selection in evolution. Oxford: Oxford University Press;
1993.
15.
Wang RS, Saadatpour A, Albert R. Boolean modeling in systems biology: an overview of methodology and
applications. Phys Biol. 2012;9(5):055001.
16.
Xiao Y. A tutorial on analysis and simulation of boolean gene regulatory network models. Curr Genomics.
2009;10(7):511–25.
17.
Rodríguez A, Sosa D, Torres L, Molina B, Frías S, Mendoza L. A Boolean network model of the FA/BRCA pathway.
Bioinformatics. 2012;28(6):858–66.
18.
Zhang R, Shah MV, Yang J, Nyland SB, Liu X, Yun JK, et al. Network model of survival signaling in large granular
lymphocyte leukemia. Proc Natl Acad Sci U S A. 2008;105(42):16308–13.
19.
Mendoza L. A network model for the control of the differentiation process in Th cells. Biosystems.
2006;84(2):101–14.
20.
Fauré A, Naldi A, Chaouiya C, Thieffry D. Dynamical analysis of a generic Boolean model for the control of the
mammalian cell cycle. Bioinformatics. 2006;22(14):e124–31.
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 13 of 15
21.
Barabási AL, Oltvai ZN. Network biology: understanding the cell’s functional organization. Nat Rev Genet.
2004;5(2):101–13.
22.
Zhang Z, Zhang J. A big world inside small-world networks. PLoS One. 2009;4(5):e5686.
23.
Freeman LC. Centrality in social networks conceptual clarification. Soc Networks. 1979;1(3):215–39.
24.
Scardoni G, Laudanna C. Centralities based analysis of complex networks. In: Zhang Y, editor. New frontiers in
graph theory. InTech; 2012. p. 323–48. Available from: http://www.intechopen.com/books/new-frontiers-in-graph-
theory/centralities-based-analysis-of-networks
25.
Newman M. A measure of betweenness centrality based on random walks. Soc Networks. 2005;27:39–54.
26.
Freeman LC. A set of measures of centrality based on betweenness. Sociometry. 1977;40(1):35–41.
27.
Yoon J, Blumer A, Lee K. An algorithm for modularity analysis of directed and weighted biological networks based
on edge-betweenness centrality. Bioinformatics. 2006;22(24):3106–8. Epub 2006 Oct 23.
28.
Assenov Y, Ramírez F, Schelhorn SE, Lengauer T, Albrecht M. Computing topological parameters of biological
networks. Bioinformatics. 2008;24(2):282–4. Epub 2007 Nov 15.
29.
Yu H, Kim PM, Sprecher E, Trifonov V, Gerstein M. The importance of bottlenecks in protein networks: correlation
with gene essentiality and expression dynamics. PLoS Comput Biol. 2007;3(4):e59. Epub 2007 Feb 14.
30.
He X, Zhang J. Why do hubs tend to be essential in protein networks? PLoS Genet. 2006;2(6):e88. Epub 2006 Apr
26.
31.
Goh KI, Cusick ME, Valle D, Childs B, Vidal M, Barabási AL. The human disease network. Proc Natl Acad Sci U S A.
2007;104(21):8685–90. Epub 2007 May 14.
32.
Klein U, Tu Y, Stolovitzky GA, Mattioli M, Cattoretti G, Husson H, et al. Gene expression profiling of B cell chronic
lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med.
2001;194(11):1625–38.
33.
Rosenwald A, Alizadeh AA, Widhopf G, Simon R, Davis RE, Yu X, et al. Relation of gene expression phenotype to
immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med.
2001;194(11):1639–47.
34.
Abruzzo LV, Barron LL, Anderson K, Newman RJ, Wierda WG, O’brien S, et al. Identification and validation of
biomarkers of IgV(H) mutation status in chronic lymphocytic leukemia using microfluidics quantitative real-time
polymerase chain reaction technology. J Mol Diagn. 2007;9(4):546–55. Epub 2007 Aug 9.
35.
Sevov M, Kaderi MA, Kanduri M, Mansouri M, Buhl AM, Cahill N. A comparative study of RNA-based markers in
chronic lymphocytic leukemia reveals LPL as a powerful predictor of clinical outcome. Haematologica.
2009;94 Suppl 3:1–95.
36.
Kaderi MA, Kanduri M, Buhl AM, Sevov M, Cahill N, Gunnarsson R, et al. LPL is the strongest prognostic factor in a
comparative analysis of RNA-based markers in early chronic lymphocytic leukemia. Haematologica.
2011;96(8):1153–60.
37.
Kolset SO, Salmivirta M. Cell surface heparan sulfate proteoglycans and lipoprotein metabolism. Cell Mol Life Sci.
1999;56(9–10):857–70.
38.
Pallasch CP, Schwamb J, Königs S, Schulz A, Debey S, Kofler D, et al. Targeting lipid metabolism by the lipoprotein
lipase inhibitor orlistat results in apoptosis of B-cell chronic lymphocytic leukemia cells. Leukemia.
2008;22(3):585–92. Epub 2007 Dec 13.
39.
Yin Y, Shen WH. PTEN: a new guardian of the genome. Oncogene. 2008;27(41):5443–53.
40.
Kanehisa M, Goto S, Sato Y, Kawashima M, Furumichi M, Tanabe M. Data, information, knowledge and principle:
back to metabolism in KEGG. Nucleic Acids Res. 2014;42(Database issue):D199–205.
41.
Pruitt K, Brown G, Tatusova T, Maglott D. The NCBI handbook [internet]. Chapter 18, the Reference Sequence
(RefSeq) project. Bethesda: National Library of Medicine (US), National Center for Biotechnology Information; 2002.
Available from http://www.ncbi.nlm.nih.gov/books/NBK21091/.
42.
Platanias LC. Map kinase signaling pathways and hematologic malignancies. Blood. 2003;101(12):4667–79. Epub
2003 Mar 6.
43.
Ferreira PG, Jares P, Rico D, Gómez-López G, Martínez-Trillos A, Villamor N, et al. Transcriptome characterization by
RNA sequencing identifies a major molecular and clinical subdivision in chronic lymphocytic leukemia. Genome
Res. 2014;24(2):212–26.
44.
Chuang HY, Rassenti L, Salcedo M, Licon K, Kohlmann A, Haferlach T, et al. Subnetwork-based analysis of chronic
lymphocytic leukemia identifies pathways that associate with disease progression. Blood. 2012;120(13):2639–49.
Epub 2012 Jul 26.
45.
Moreau Y, Aerts S, De Moor B, De Strooper B, Dabrowski M. Comparison and meta-analysis of microarray data:
from the bench to the computer desk. Trends Genet. 2003;19(10):570–7.
46.
Huber W, von Heydebreck A, Sültmann H, Poustka A, Vingron M. Variance stabilization applied to microarray data
calibration and to the quantification of differential expression. Bioinformatics. 2002;18 Suppl 1:S96–104.
47.
Wang X, Kang DD, Shen K, Song C, Lu S, Chang LC, et al. An R package suite for microarray meta-analysis in
quality control, differentially expressed gene analysis and pathway enrichment detection. Bioinformatics.
2012;28(19):2534–6. Epub 2012 Aug 3.
48.
Rhodes DR, Barrette TR, Rubin MA, Ghosh D, Chinnaiyan AM. Meta-analysis of microarrays: interstudy validation of
gene expression profiles reveals pathway dysregulation in prostate cancer. Cancer Res. 2002;62(15):4427–33.
49.
Jensen LJ, Kuhn M, Stark M, Chaffron S, Creevey C, Muller J, et al. STRING 8—a global view on proteins and their
functional interactions in 630 organisms. Nucleic Acids Res. 2009;37(Database issue):D412–6.
50.
Szklarczyk D, Franceschini A, Kuhn M, Simonovic M, Roth A, Minguez P, et al. The STRING database in 2011:
functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res. 2011;39(Database
issue):D561–8.
51.
Bader GD, Betel D, Hogue CW. BIND: the Biomolecular Interaction Network Database. Nucleic Acids Res.
2003;31(1):248–50.
52.
Breitkreutz BJ, Stark C, Reguly T, Boucher L, Breitkreutz A, Livstone M, et al. The BioGRID interaction database: 2008
update. Nucleic Acids Res. 2008;36(Database issue):D637–40. Epub 2007 Nov 13.
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 14 of 15
53.
Xenarios I, Rice DW, Salwinski L, Baron MK, Marcotte EM, Eisenberg D. DIP: the database of interacting proteins.
Nucleic Acids Res. 2000;28(1):289–91.
54.
Orchard S, Ammari M, Aranda B, Breuza L, Briganti L, Broackes-Carter F, et al. The MIntAct project–IntAct as a common
curation platform for 11 molecular interaction databases. Nucleic Acids Res. 2014;42(Database issue):D358–63.
55.
Keshava Prasad TS, Goel R, Kandasamy K, Keerthikumar S, Kumar S, Mathivanan S, et al. Human protein reference
database–2009 update. Nucleic Acids Res. 2009;37(Database issue):D767–72.
56.
Schaefer CF, Anthony K, Krupa S, Buchoff J, Day M, Hannay T, et al. PID: the pathway interaction database. Nucleic
Acids Res. 2009;37(Database issue):D674–9.
57.
Croft D, O’Kelly G, Wu G, Haw R, Gillespie M, Matthews L, et al. Reactome: a database of reactions, pathways and
biological processes. Nucleic Acids Res. 2011;39(Database issue):D691–7.
58.
Kanehisa M, Goto S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 2000;28(1):27–30.
59.
Klingström T, Plewczynski D. Protein-protein interaction and pathway databases, a graphical review. Brief Bioinform.
2011;12(6):702–13.
60.
Doncheva NT, Assenov Y, Domingues FS, Albrecht M. Topological analysis and interactive visualization of
biological networks and protein structures. Nat Protoc. 2012;7(4):670–85.
61.
Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for
integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.
62.
Scardoni G, Petterlini M, Laudanna C. Analyzing biological network parameters with CentiScaPe. Bioinformatics.
2009;25(21):2857–9.
63.
Albert I, Thakar J, Li S, Zhang R, Albert R. Boolean network simulations for life scientists. Source Code Biol Med.
2008;3:16.
64.
Fumiã HF, Martins ML. Boolean network model for cancer pathways: predicting carcinogenesis and targeted
therapy outcomes. PLoS One. 2013;8(7), e69008.
65.
Müssel C, Hopfensitz M, Kestler HA. BoolNet–an R package for generation, reconstruction and analysis of Boolean
networks. Bioinformatics. 2010;26(10):1378–80.
66.
Tabas-Madrid D, Nogales-Cadenas R, Pascual-Montano A. GeneCodis3: a non-redundant and modular enrichment
analysis tool for functional genomics. Nucleic Acids Res. 2012;40(Web Server issue):W478–83.
67.
Nogales-Cadenas R, Carmona-Saez P, Vazquez M, Vicente C, Yang X, Tirado F, et al. GeneCodis: interpreting gene
lists through enrichment analysis and integration of diverse biological information. Nucleic Acids Res.
2009;37(Web Server issue):W317–22.
68.
Carmona-Saez P, Chagoyen M, Tirado F, Carazo JM, Pascual-Montano A. GENECODIS: a web-based tool for finding
significant concurrent annotations in gene lists. Genome Biol. 2007;8(1):R3.
Submit your next manuscript to BioMed Central
and take full advantage of:
• Convenient online submission
• Thorough peer review
• No space constraints or color figure charges
• Immediate publication on acceptance
• Inclusion in PubMed, CAS, Scopus and Google Scholar
• Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Álvarez-Silva et al. Theoretical Biology and Medical Modelling (2015) 12:12
Page 15 of 15
|
26088082
|
MAP2K6 = ( CCL5 )
ILK = NOT ( ( FOS ) )
RUNX3 = NOT ( ( EZH2 ) )
HSP90AA1 = ( HSP90AA1 )
CCND2 = ( ( ANXA2 ) AND NOT ( PTEN ) )
CD81 = ( CD81 )
PTEN = ( ( ( ( ( ( FRK AND ( ( ( INPP5D AND BMI1 AND ILK AND IFNGR2 AND RRM1 AND EGR1 AND TNF ) ) ) ) AND NOT ( ADM ) ) AND NOT ( BMI1 ) ) AND NOT ( TGFBR3 ) ) AND NOT ( CSNK2A2 ) ) AND NOT ( AEBP1 ) ) OR ( ( ( ( EGR1 AND ( ( ( FRK AND INPP5D AND BMI1 AND ILK AND RRM1 ) ) ) ) AND NOT ( BMI1 ) ) AND NOT ( TGFBR3 ) ) AND NOT ( AEBP1 ) )
BLNK = ( CD79A )
MAP4K4 = ( TNF )
AICDA = ( HSP90AA1 ) OR ( ( CD27 AND ( ( ( HSP90AA1 ) ) ) ) AND NOT ( CD79A ) )
CREM = ( CREM )
TNF = ( LPL )
FGFR1 = ( FGFR1 )
CHST2 = ( CHST2 )
CD79A = ( TCF3 AND ( ( ( CD86 ) ) ) )
TNFRSF1B = ( TNFRSF1B )
LIG1 = ( PCNA )
DPP4 = ( TNF )
FCER2 = ( NOTCH2 )
CD86 = NOT ( ( TNFRSF1B ) )
AKAP12 = ( TNF )
CD70 = ( TNF )
FOS = ( ( ADM AND ( ( ( LMNA AND MAP2K6 AND TNF ) ) ) ) AND NOT ( JAK1 ) ) OR ( ( ( CNR1 AND ( ( ( LMNA AND MAP2K6 AND ADM AND PCNA AND TNF ) ) ) ) AND NOT ( CREM ) ) AND NOT ( JAK1 ) )
AKT3 = ( IGF1R AND ( ( ( NOT PTEN ) ) ) )
GSK3B = ( GSK3B )
FRK = ( FRK )
OGT = ( IGF1R )
NFE2L2 = ( ( TNF ) AND NOT ( GSK3B ) )
CAV1 = ( PRKCA ) OR ( TGFBR3 AND ( ( ( PRKCA ) ) ) )
APLP2 = ( APP )
HSP90B1 = NOT ( ( IFNGR2 ) )
SIAH1 = ( SIAH1 )
NOTCH2 = ( TNF )
HDAC9 = ( ( NCOR2 ) AND NOT ( SKI ) )
MIF = ( CD74 AND ( ( ( IFNGR2 AND TNF ) ) ) )
CD27 = NOT ( ( PRF1 ) )
RPS6KA5 = ( TNF )
PAX3 = ( ( PTCH1 ) AND NOT ( TGFBR3 ) )
EED = ( EED )
PTCH1 = ( FGFR1 )
APP = ( ( ( FYN ) AND NOT ( CD74 ) ) AND NOT ( CD74 ) ) OR ( IFNGR2 AND ( ( ( TNF ) ) ) )
AEBP1 = ( TGFBR3 )
H2AFX = ( CASP3 )
BMI1 = ( BMI1 )
EZH2 = ( ( ( EED ) AND NOT ( HDAC9 ) ) ) OR NOT ( EED OR HDAC9 )
CUL5 = ( CUL5 )
PLD1 = ( PRKCA AND ( ( ( APP ) ) ) )
MYLK = ( TNF )
JAK1 = ( ( IFNGR2 AND ( ( ( IL7 AND IL10RA ) ) ) ) AND NOT ( HDAC9 ) )
PRKCA = ( TGFBR3 AND ( ( ( AKAP12 ) ) ) )
HIST2H2AA3 = NOT ( ( HDAC9 ) )
CCL5 = ( ( IFNGR2 ) AND NOT ( FOS ) )
LMNA = ( LMNA )
IL10RA = ( IL10RA )
AFF1 = NOT ( ( SIAH1 ) )
RFC5 = ( RFC5 )
CNR1 = ( CNR1 )
ALOX5 = ( TGFBR3 AND ( ( ( EGR1 ) ) ) ) OR ( EGR1 )
TCF3 = ( TCF3 )
RRM1 = ( RRM1 )
SELL = ( CHST2 AND ( ( ( IFNGR2 ) ) ) )
FYN = ( FYN )
CD63 = ( SELP )
MYL9 = ( MYLK )
CD74 = ( CD74 )
ANXA2 = ( ANXA2 )
HIST1H3H = NOT ( ( HIST2H2AA3 ) )
LPL = ( ( FOS ) AND NOT ( TNF ) )
PRF1 = ( TNF AND ( ( ( RUNX3 ) ) ) ) OR ( RUNX3 )
CD69 = ( TNF AND ( ( ( CD81 ) ) ) )
ADM = ( HSP90AA1 ) OR ( TNF AND ( ( ( HSP90AA1 ) ) ) )
IFNGR2 = ( TNF )
PCNA = ( ( CSDA AND ( ( ( HSP90AA1 ) ) ) ) AND NOT ( RFC5 ) )
MARCKS = NOT ( ( PRKCA ) )
LGALS1 = ( TGFBR3 )
IL7 = ( IFNGR2 AND ( ( ( TNF ) ) ) ) OR ( TNF )
VDR = ( ( PRKCA ) AND NOT ( NCOR2 ) )
SELP = ( MGAT5 AND ( ( ( TNF ) ) ) )
CSDA = ( CSDA )
MGAT5 = NOT ( ( LGALS1 ) )
CTSB = ( CAV1 )
APOBEC3G = ( ( ( IFNGR2 ) AND NOT ( CUL5 ) ) ) OR NOT ( CUL5 OR IFNGR2 )
IGF1R = ( EGR1 AND ( ( ( CAV1 ) ) ) )
NCOR2 = ( TNF )
CASP3 = ( TNF AND ( ( ( NOT CTSB AND NOT IGF1R ) ) ) )
NAB1 = ( EGR1 )
SKI = ( SKI )
CSNK2A2 = ( CSNK2A2 )
TGFBR3 = ( TGFBR3 )
EGR1 = ( ( ( CNR1 AND ( ( ( TGFBR3 AND AEBP1 ) ) ) ) AND NOT ( NAB1 ) ) AND NOT ( HDAC9 ) )
INPP5D = ( TGFBR3 ) OR ( IGF1R AND ( ( ( TGFBR3 ) ) ) )
|
RESEARCH ARTICLE
A Minimal Regulatory Network of Extrinsic
and Intrinsic Factors Recovers Observed
Patterns of CD4+ T Cell Differentiation and
Plasticity
Mariana Esther Martinez-Sanchez1,2, Luis Mendoza3, Carlos Villarreal2,4, Elena R. Alvarez-
Buylla1,2*
1 Departamento de Ecología Funcional, Instituto de Ecología, Universidad Nacional Autónoma de México,
Coyoacán, México Distrito Federal, México, 2 Centro de Ciencias de la Complejidad, Universidad Nacional
Autónoma de México, Coyoacán, México Distrito Federal, México, 3 Departamento de Biología Molecular y
Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México
Distrito Federal, México, 4 Departamento de Física Teórica, Instituto de Física, Universidad Nacional
Autónoma de México, México Distrito Federal, México
* eabuylla@gmail.com
Abstract
CD4+ T cells orchestrate the adaptive immune response in vertebrates. While both experi-
mental and modeling work has been conducted to understand the molecular genetic mech-
anisms involved in CD4+ T cell responses and fate attainment, the dynamic role of intrinsic
(produced by CD4+ T lymphocytes) versus extrinsic (produced by other cells) components
remains unclear, and the mechanistic and dynamic understanding of the plastic responses
of these cells remains incomplete. In this work, we studied a regulatory network for the core
transcription factors involved in CD4+ T cell-fate attainment. We first show that this core is
not sufficient to recover common CD4+ T phenotypes. We thus postulate a minimal Boolean
regulatory network model derived from a larger and more comprehensive network that is
based on experimental data. The minimal network integrates transcriptional regulation, sig-
naling pathways and the micro-environment. This network model recovers reported configu-
rations of most of the characterized cell types (Th0, Th1, Th2, Th17, Tfh, Th9, iTreg, and
Foxp3-independent T regulatory cells). This transcriptional-signaling regulatory network is
robust and recovers mutant configurations that have been reported experimentally. Addi-
tionally, this model recovers many of the plasticity patterns documented for different T CD4
+ cell types, as summarized in a cell-fate map. We tested the effects of various micro-envi-
ronments and transient perturbations on such transitions among CD4+ T cell types. Inter-
estingly, most cell-fate transitions were induced by transient activations, with the opposite
behavior associated with transient inhibitions. Finally, we used a novel methodology was
used to establish that T-bet, TGF-β and suppressors of cytokine signaling proteins are
keys to recovering observed CD4+ T cell plastic responses. In conclusion, the observed
CD4+ T cell-types and transition patterns emerge from the feedback between the intrinsic
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
1 / 23
OPEN ACCESS
Citation: Martinez-Sanchez ME, Mendoza L,
Villarreal C, Alvarez-Buylla ER (2015) A Minimal
Regulatory Network of Extrinsic and Intrinsic Factors
Recovers Observed Patterns of CD4+ T Cell
Differentiation and Plasticity. PLoS Comput Biol 11(6):
e1004324. doi:10.1371/journal.pcbi.1004324
Editor: Josep Bassaganya-Riera, Virginia Tech,
UNITED STATES
Received: December 5, 2014
Accepted: May 7, 2015
Published: June 19, 2015
Copyright: © 2015 Martinez-Sanchez et al. This is
an open access article distributed under the terms of
the Creative Commons Attribution License, which
permits unrestricted use, distribution, and
reproduction in any medium, provided the original
author and source are credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Additionally, the models presented can be found at
BioModels Database (acession numbers:
MODEL1411170000 and MODEL1411170001). URL:
https://www.ebi.ac.uk/biomodels/reviews/
MODEL1411170000-1/
Funding: This work was supported by grants from:
Consejo Nacional de Ciencia y Tecnología México
(www.conacyt.mx): 180380 to CV and ERAB, and
180098 to (ERAB). Programa de Apoyo a Proyectos
de Investigación e Innovación Tecnológica
or intracellular regulatory core and the micro-environment. We discuss the broader use of
this approach for other plastic systems and possible therapeutic interventions.
Author Summary
CD4+ T cells orchestrate adaptive immune responses in vertebrates. These cells differenti-
ate into several types depending on environmental signals and immunological challenges.
Once these cells are committed to a particular fate, they can switch to different cell types,
thus exhibiting plasticity that enables the immune system to dynamically adapt to novel
challenges. We integrated available experimental data into a large network that was for-
mally reduced to a minimal regulatory module with a sufficient set of components and in-
teractions to recover most CD4+ T cell types and reported plasticity patterns in response
to various micro-environments and transient perturbations. We formally demonstrate
that transcriptional regulatory interactions are not sufficient to recover CD4+ T cell types
and thus propose a minimal network that induces most observed phenotypes. This model
is robust and was validated with mutant CD4+ T phenotypes. The model was also used to
identify key components for cell differentiation and plasticity under varying immunogenic
conditions. The model presented here may be a useful framework to study other plastic
systems and guide therapeutic approaches to immune system modulation.
Introduction
The immune system protects organisms against external agents that may cause various types of
diseases. As the immune system mounts specialized responses to diverse pathogens, it relies on
plastic responses to changing immunological challenges. At the same time, the immune system
must maintain homeostasis and avoid auto-immune responses. Therefore, the immune system
relies on resilience mechanisms that enable it to return to basal conditions once pathogens or
immunogenic factors are no longer present [1–3].
CD4+ T cells, also known as T helper (Th) cells, are key in the response to infectious agents
and in the plasticity of the immune system. Naive CD4+ T cells (Th0) are activated when they
recognize an antigen in a secondary lymphoid organ. Depending on the cytokine milieu and
other signals in their micro-environment, CD4+ T cells attain different cell fates [2,4–7]. None-
theless, we still do not have a complete understanding of the dynamic mechanisms underlying
CD4+ T cell differentiation and plasticity [5].
Each CD4+ T cell type is associated with specific cytokines, receptors, transcription factors
and functions (Fig 1). Th1 cells express T-bet, secrete interferon-γ (IFN-γ) and are associated
with cellular immunity [8]. Th2 cells express GATA3, secrete interleukin (IL)-4 and are associ-
ated with immunity to parasites [8]. Th17 cells express RORα and RORγt, secrete IL-17 and
IL-21, and are associated with neutrophil activation [9–10]. Follicular helper CD4+ T cells
(Tfh) express Bcl6 and CXCR5, secrete IL-21 and are associated with B cell maturation in ger-
minal centers [11,12]. Th9 cells secrete IL-9 and exert anticancer activity [13,14]. Induced regu-
latory T cells express Foxp3, secrete TGF-β and/or IL-10, and are associated with immune
tolerance [15,16]. There is also considerable overlap among the expression profiles of different
CD4+ T cells. For example, IL-9 and IL-10 can be secreted by Th1, Th2, Th17, iTreg cells and a
variety of other immune cells [17–19]. T regulatory cells can also express IL-17 [20].
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
2 / 23
Universidad Nacional Autónoma de México (dgapa.
unam.mx/html/papiit/papit.html): IN203113; IN204011;
IN226510-3 and IN203814 to ERAB. Programa de
Apoyo a Proyectos de Investigación e Innovación
Tecnológica Universidad Nacional Autónoma de
México (dgapa.unam.mx/html/papiit/papit.html):
IN200514 to LM. MEMS acknowledges support from
the graduate program “Doctorado en Ciencias
Biomédicas, de la Universidad Nacional Autónoma
de México”. The funders had no role in study design,
data collection and analysis, decision to publish, or
preparation of the manuscript.
Competing Interests: The authors have declared
that no competing interests exist.
CD4+ T cells are highly plastic, switching from one type to another in response to environ-
mental challenges (Fig 1) [1,21–23]. Th17 cells can transform into Th1 cells [24–25], and
iTregs differentiate into Th17 in the presence of IL-6 [26]. Th2 cells can become IL-9 produc-
ing cells but may not easily become Th1 cells [27]. iTreg and Tfh cells can independently devel-
op into other CD4+ T cell types, and they can be derived from Th1, Th2 or Th17 cells [28–30].
The differentiation and plasticity of CD4+ T cells depends on the interactions among the cyto-
kines produced by other immune cells, epithelial cells, adipocytes, or by the CD4+ T cells
themselves; the transduction of those signals and the regulation of this signaling by suppressors
of cytokine signalling (SOCS) proteins; the set of transcription factors expressed inside the
cells; epigenetic regulation; certain metabolites; and also microRNAs [4,6,31–33]. Given the
Fig 1. Differentiation and plasticity of CD4+ T cell types. CD4+ T cell types are characterized by their unique cytokine production profiles, transcription
factors and biological functions. The main cell types are Th0, Th1, Th2, Th17, iTreg and Tfh. Other possible cell types have been described such as IL-9
(Th9), IL-10+Foxp3-(Tr1) and TGF-β+Foxp3-(Th3) producing cells.
doi:10.1371/journal.pcbi.1004324.g001
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
3 / 23
complexity of CD4+ T cell transitions and the difficulty of classifying a particular expression
pattern as a subset or a lineage, we will refer to the different stable expression patterns of CD4
+ T cells as “cell-types”.
A mechanistic, integrative and system-level understanding of CD4+ T cell differentiation
and plasticity requires dynamic regulatory network models that consider the concerted action
of many components. These models can be used to prove whether the known biological inter-
actions are necessary and sufficient to recover attractors that correspond to experimentally ob-
served configurations in different CD4+ T cell types. Additionally, such models may be used to
address whether the considered components and interactions also restrict and explain the ob-
served patterns of transition among cell types. Finally, this type of model can be used to test the
role of different network components in cell differentiation and plasticity.
In such regulatory network models, the nodes correspond to the regulatory components of
the network such as genes, proteins or signals, while the links correspond to the interactions
among components. The state of each node is determined by the expression level of its regula-
tors, and the logical functions describe the dynamic evolution of the node states. The attractors,
the states to which such regulatory networks converge, can be interpreted as the profiles char-
acterizing different cell types (see reviews in: [34–36]).
Previous studies have used regulatory network models to study CD4+ T cell differentiation
and plasticity [37–40]. These models captured the dynamic and non-linear regulation of CD4
+ T cells and recovered the attractors corresponding to the Th0, Th1, Th2, iTreg and Th17 cell
types. They have also been useful for preliminary studies of CD4+ T cell plasticity in the pres-
ence of different cytokines in the micro-environment [38] and fir studies of the effect of a spe-
cific molecule (PPARγ) in the Th17/iTreg switch [40]. However, as new T CD4+ cell types
such as Tfh, regulatory Foxp3-independent, Th9, and Th22 cells are described, it is necessary
to develop an updated regulatory network that is able to recover the configurations that charac-
terize such novel cell subsets. Additionally, to date no minimal model that incorporates the
necessary and sufficient set of interactions to also recover the reported patterns of transitions
among Th cells has been reported.
Here, we specifically address whether CD4+ cell types and their transition patterns emerge
as a result of the feedback between a minimal regulatory core of intra-cellular transcription fac-
tors and cytokines produced by the CD4+ T cell together with cytokines produced by other
cells present in the micro-environment. Our results confirm that a regulatory network model
that only considers the interactions among the master transcription factors is not sufficient to
recover configurations that characterize the different CD4+ T cell types. Therefore, we then in-
tegrated a minimal network of master transcriptional factors with cytokine signaling pathway,
including the cytokines produced by the cell and those present in the micro-environment, to
integrate a network with the necessary and sufficient set of components to recover documented
CD4+ T cell differentiation and plasticity patterns. The observed configurations of CD4+ T
cells (Th0, Th1, Th2, Th17, iTreg, Tfh, Th9 and Foxp3-independent T regulatory cells) emerge
from the feedback and cooperative dynamics among the multiple levels of regulation consid-
ered in the minimal model. In addition, this system is able to recover the plastic transition pat-
terns and stability behavior that have been described for the different cell types in response to
transitory perturbations and different micro-environments. Interestingly, our model predicts
that transitions from particular cell types to others are caused by transient activations, while
transient inhibitions usually cause cells to remain in their original cell types. Additionally, we
show that T-bet, TGF-β and SOCS proteins are keys to recovering observed CD4+ T cell plastic
responses. Finally, we discuss the relevance of our models for a system-level understanding of
mammalian immunological responses and eventual biomedical interventions.
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
4 / 23
Results
CD4+ T cell regulatory network
Boolean networks are capable of integrating qualitative interactions (molecular, physical,
chemical, etc.) into a coherent picture and are useful ways to explore the minimal set of restric-
tions that are necessary and sufficient to produce emergent biological patterns and behaviors
[41–43]. The regulatory interactions considered in the present model are grounded on experi-
mental data. In the proposed regulatory network, the nodes represent the regulatory compo-
nents of the network and the links the interactions among them (S1 Table and S1 Fig). Given
the complexity of the network, we simplified the model by removing intermediate components
along a network path (S1 File) following a method proposed in [44] and checked the consisten-
cy of the reduced network using GINsim [45].
The predicted cell phenotypes arising from the steady states of the network are consistent
with the available experimental data [2,4–7]. The model assumes that all interactions are syn-
chronous, that all cytokine receptors are present, and that the TCR and its cofactors are activat-
ed (being unable to model unactivated and anergic CD4+ T cells). The model ignores weak
interactions, low levels of expression, and epigenetic regulation (S1 File).
A core of master transcriptional regulators is not sufficient to explain
CD4+ T cell differentiation
To address whether a minimal transcriptional regulatory core could recover the observed con-
figurations that characterize the main CD4+ T cell types that have been described up to now,
we extracted from the general network under study a minimal regulatory module consisting
only of transcriptional regulators (Fig 2A, S2 Table, BioModels Database:
MODEL1411170000). Our aim was to test whether this minimal module contained a sufficient
set of interactions to predict the observed configurations for the transcription factors included
in the model that characterize different CD4+ T cell types. The nodes of the transcriptional
Fig 2. Minimal network of master transcriptional regulators CD4+ T (CD4+ T TRN). Based on published
experimental data we constructed a CD4+ T cell regulatory network that includes the master transcriptional
regulators and the interactions among those regulators (CD4+ T TRN). (A) Graph of the CD4+ T TRN. Node
colors correspond to cell types: Th1 (yellow), Th2 (green), Th17 (red), iTreg (blue) and Tfh (purple).
Activations among elements are represented with black arrows and inhibitions with red dotted arrows. (B)
Attractors of the CD4+ T TRN: Each column corresponds to an attractor. Each node can be active (green) or
inactive (red). The attractors correspond to configurations that characterize the Th0, Th1, Th2, iTreg, T-bet
+Foxp3+ and GATA3+Foxp3+ types. The attractors corresponding to the Th17 and Tfh types could not
be recovered.
doi:10.1371/journal.pcbi.1004324.g002
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
5 / 23
regulatory network (TRN) correspond to the five “master” transcription factors associated with
CD4+ T cell types: T-bet for Th1, GATA3 for Th2, RORγt for Th17, Foxp3 for iTreg, and Bcl6
for Tfh.
The dynamic analysis of this TRN recovered attractors corresponding to different CD4+ T
cell types (Fig 2B): Th0, Th1, Th2, iTreg and the hybrid states T-bet+Foxp3+ [46] and GATA3
+Foxp3+ [47]. However, this TRN did not converge to configurations that characterize the
Th17 and Tfh cell types, implying that the expression of RORγt and Bcl6 is not sufficient to
maintain such cell types. This result may be caused by the lack of feed-forward loops in the
TRN. RORγt has no positive interactions with any of the transcription factors considered in
the TRN and lacks a feedback loop mediated by transcription factors [48]. The mode of self-
regulation of Bcl6 remains unclear, as it has been reported to either activate or inhibit its own
expression in B cells [49–50].
CD4+ T cell differentiation patterns emerge from feedback between the
transcriptional regulatory network, cytokines and signaling pathways
The above result reveals which T CD4+ cell types rely only on the postulated TRN and which
require extrinsic signals. To formally test this hypothesis, we extended the TRN network by in-
troducing key components of signaling pathways and their regulators, as well as cytokines that
have been shown to be fundamental in CD4+ T cell type attainment. This T CD4+ cell tran-
scriptional-signaling regulatory network (TSRN) was then simplified (S1 File, S1 Fig) to obtain
a minimal network. To reduce the number of nodes in the network, we assumed that the TCR
signal was present and that the cytokine receptors were present in sufficient amounts to trans-
duce a signal. This network lacks many important inflammatory cytokines (such as IL-1,
TNFα), because while these cytokines are crucial for the immune response, they are dispens-
able for CD4+ T cell differentiation. The model analyzed in this paper also lacks extrinsic cyto-
kines produced by other immune system cells and other cell types such as IL-12 and IL-18. The
network also lacks some transcription factors and cytokines associated with newly reported Th
types such as IL-22, as detailed experimental information linking them to the network model
under analysis is not yet available.
The nodes of the simplified TSRN represent (Fig 3A, S3 Table, BioModels Database:
MODEL1411170001) transcription factors, signaling pathways and extrinsic cytokines. The
nodes corresponding to cytokine pathways are active if the signal is transduced; this means
that if the cytokine is present, it forms a complex with the receptor that can activate a messen-
ger molecule (for example a STAT protein), which is then translocated to the nucleus. Cyto-
kines can be produced by both CD4+ T cells (intrinsic) and by other cells of the immune
system and the organism (extrinsic). To resolve this ambiguity we added nodes representing
the extrinsic cytokines produced by other cells and tissues of the immune system (IL_e). This
extended TSRN includes 18 nodes: the transcription factors (Tbet, GATA3, RORγt, Foxp3,
Bcl6), the effector cytokines and their signaling pathways (IFN-γ, IL-2, IL-4, IL-21, IL-9), the
regulatory cytokines (TGF-β and IL-10) and the extrinsic cytokines (IFN-γe, IL-2e, IL-4e, IL-
21e, TGF-βe and IL-10e). While IL-10, IL-6 and IL-21 all signal using STAT3, IL-6 and IL-21
cause inflammation, while IL-10 suppresses inflammation. To analyze this network, we assume
that IL-10 signaling was mediated by a different pathway than IL-6/IL-21, even though they
share STAT3 as a messenger molecule. The production of these external cytokines is indepen-
dent of regulation inside the CD4+ T cell, but their signaling can be blocked (for example by
SOCS proteins [51]). The resulting network includes two levels of regulation, the regulation in
the nucleus by mutually inhibiting transcription factors and the regulation among the receptors
and their signal transduction pathways mediated by SOCS proteins.
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
6 / 23
Fig 3. CD4+ T cell transcriptional-signaling regulatory network (TSRN). We constructed a regulatory
network using available experimental data. The network includes transcription factors, signaling pathways,
and intrinsic and extrinsic cytokines. (A) Graph of the TSRN. The nodes include transcription factors
(rectangles), intrinsic cytokines and their signaling pathways (ellipses) and extrinsic cytokines (ellipses).
Node colors correspond to cell type: Th1 (yellow), Th2 (green), Th17 (red), iTreg (blue), Tfh (purple), and Th9
(brown). Activations between elements are represented with black arrows, and inhibitions with red dotted
arrows. The dotted lines represent inhibition mediated by SOCS proteins. (B) Attractors of the TSRN. Each
column corresponds to an attractor. Each node can be active (green) or inactive (red), extrinsic cytokines
may be active or inactive (yellow). The following attractors were found in the network: Th0, Th1, Th2, Th17,
iTreg, Tfh, Th9 producing T cells, Foxp3-independent T regulatory cells (TrFoxp3-), T-bet+ T regulatory cells
(Th1R), GATA3+ T regulatory cells (Th2R) and GATA3+IL-4- cells. Attractors where labeled according to the
active transcription factors and intrinsic cytokines.
doi:10.1371/journal.pcbi.1004324.g003
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
7 / 23
The dynamic analysis of the TSRN yields stable configurations that correspond to: Th0,
Th1, Th2, Th17, iTreg, Tfh, T regulatory Foxp3-independent cells, Th1R, Th2R and GATA3
+IL4- cells (Fig 3B). As this biological patterns can be obtained in the presence of various ex-
trinsic cytokines, we labeled each attractor according to the active transcription factors and
intrinsic cytokines. Resting CD4+ T cells (labeled Th0) were defined as expressing no tran-
scription factors or regulatory cytokines. Th1 was defined as Tbet and IFN-γ active [8], Th2 as
GATA3 and IL-4 active [8] and GATA3+ (a Th2-like cell type) as GATA3+IL4-[38]. Th17 was
defined based on RORγt and STAT3 signaling mediated by IL-6 or IL-21, all of which require
the presence of TGF-βe [9–10]. iTreg expressed Foxp3 and TGF-β, IL-10 or both, all of which
require the presence of IL-2e [16]. Interestingly, the TSRN model also predicts a novel set of
steady states that had not been predicted by previous models but that correspond to reported
biological cell types (Fig 3B); for example, Tfh cells with Bcl6 and STAT3 signaling mediated
by IL-21 [12]; Th9 cells with IL-9, requiring the presence of TGF-β and extrinsic IL-4 [27]; T
regulatory cells, as Foxp3-independent CD4+ T cells (TrFoxp3-) with TGF-β, IL-10 or both,
but not Foxp3 [52]; Th1 regulatory cells (Th1R) expressing a regulatory cytokine and T-bet
[46]; and Th2 regulatory cells (Th2rR) expressing a regulatory cytokine and GATA3 [47]. The
model does not consider the Th22 cell type [53] because IL-22 was not included in the network
due to the lack of experimental data on this molecule.
To validate the model with experimental data, we simulated loss and gain of function alter-
ations for some nodes. In general, the results agree with the available experimental data, except
in the case of the IL-2 knock-out. IL-2- causes the loss of iTreg cells as these cells require con-
tinuous IL-2 signaling [54,55], but this differs from the actual IL-2 KO mutants, which lose
most CD4+ T cell types because IL-2 is also critical for the activation and survival of CD4+ T
cells. This model also allows us to predict the behavior of the Tr Foxp3-, Th1R and Th2R cell
types in response to various knock-out and over-expression simulations for several transcrip-
tion factors or signaling pathways where no experimental data are available.
We performed a functional robustness analysis in which the logical functions of the network
were altered (S2 Fig) to verify the construction of the functions and the structural properties of
the model and to avoid over-fitting. Altering one of the functions of the network resulted in
1.389% of the initial states attaining a different final attractor than the original final state, and
only 0.219% of the initial states arrived at an attractor that was not in the original set of attrac-
tors of the non-altered network.
To further verify that the results of the Boolean network are not an artifact due to the dis-
crete nature of the model and to further assess the robustness of the attractors to variations in
the node values, we approximated the discrete step-like functions of the Boolean model with
continuous interaction functions [44] (S2 File). The continuous model recovers the same at-
tractors as the Boolean regulatory network. Furthermore, these attractors are stable in response
to small perturbations in the value of the nodes as predicted by the robustness analyses of the
Boolean version of the model.
CD4+ T cell differentiation in response to the micro-environment
Cytokines can be produced by the cell (intrinsic) or by other cells of the immune system (ex-
trinsic). These extrinsic cytokines constitute the micro-environment for CD4+ T cell differenti-
ation. The role of polarizing micro-environments in CD4+ T cell differentiation was assessed
using the TSRN model. In this network, the values of the extrinsic cytokines were fixed at a
given expression level and the network response was analyzed again (Fig 4). Th0, Th1, Th2 and
Tfh can be maintained in the absence of extrinsic cytokines or in the presence of effector cyto-
kines such as IFN-γ, IL-2, IL-4 or IL-21. Th17, iTreg and Th9 cells require extrinsic TGF-β, IL-
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
8 / 23
2 and IL-4, respectively, to maintain their homeostatic states [13,56]. TrFoxp3- states can be
maintained in most polarizing micro-environments [57,58]. The recovered behaviors agree
with the experimental data and also with previous models [38].
The importance of the extrinsic cytokines present in the micro-environment can be further
analyzed when the system is studied under polarizing conditions. The presence of extrinsic sig-
nals for a given cell type increases the number of initial states that differentiate into that cell
type, while the absence of extrinsic signals may lead to the loss of a cell type, as is the case with
Th17, iTreg and Th9 cells (Fig 4). The presence of the regulatory cytokines IL-10 and TGF-β
inhibits most effector CD4+ T cells, except for Th17. This finding may explain the presence of
Th17 cells in regulatory micro-environments [59] and provides important insight concerning
the relationship between Th17 and iTreg. Thus, this type of modeling framework and analysis
may prove useful for finding therapeutic approaches to chronic inflammation.
The polarization of the micro-environment towards a particular cell type increases the size of
the basin of attraction and its resistance to transient perturbations. Basin size and attractor stabil-
ity are not identical (S3 Fig). In this way, the environmental signals promote specific cell types
and increase their stability, which likely affects the population dynamics of CD4+ T cells. None-
theless, different CD4+ T cell types coexist during immune responses. Even if the signals in the
micro-environment promote a specific cell type, attractors corresponding to other cell types can
still appear in this micro-environment, but their basin sizes and stability tend to be smaller.
CD4+ T cell plasticity in response to the micro-environment
The ability of the immune system to dynamically respond to environmental challenges de-
pends on its plastic responses. CD4+ T cells are phenotypically plastic, and once differentiated,
Fig 4. Effect of the micro-environment on CD4+ T cell differentiation as determined using the TSRN
model. The values of the extrinsic signals of the TSRN were fixed according to different polarizing micro-
environments. The basins of attraction of the resulting attractors were plotted on a logarithmic scale. The
following micro-environments were studied: combinations of all extrinsic cytokines, no extrinsic cytokines
(Th0), IFN-γe (Th1), IL-4e and IL-2e (Th2), IL-21e and TGF-βe (Th17), TGF-βe and IL-2e (iTreg), IL-10e
(IL10), IL-21e (Tfh), and IL-4e and TGF-βe (Th9).
doi:10.1371/journal.pcbi.1004324.g004
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
9 / 23
their expression patterns can be altered depending on internal and external cues. This cell plas-
ticity seems to be important for the overall plasticity of immune system responses [1].
To analyze CD4+ T cell plasticity, we transiently perturbed the attractors of the system. For
each attractor we altered the value of one of its nodes and then evaluated the system until an at-
tractor was reached. If the original attractor was reached, we considered the corresponding cell
type as stable towards that perturbation. If a new cell-type was reached, we considered that the
transition from one cell type to another corresponded to phenotypic plasticity. This analysis
was repeated for every node and every attractor. This methodology allowed us determine all
the transitions between cell types, the specific perturbation that caused the transition, and the
path from one cell-type to another. These transient perturbations in the values of the nodes are
equivalent to developmental noise or temporal changes in the micro-environment of the cell.
The result is a cell-fate map where the nodes represent CD4+ T cell types recovered by the
TSRNand the connections represent the possible transitions between pairs of differentiated cell
types (Fig 5, S3 File).
The model recovers the reported transitions corresponding to the polarization of naïve CD4
+ T cells into canonical CD4+ T cell types, as well as various events of trans-differentiation be-
tween canonical CD4+ T cell types. Most of the predicted transitions are to or from Th0 or to-
wards TrFoxp3-. It is important to clarify that the TCR complex was not included in the
minimal model. Thus, in our model, the Th0 attractor represents resting CD4+ T cells. There
are few direct transitions among the Th1, Th2, and Th17 cell types. The few direct transitions
found towards iTreg and Tfh can only be achieved in polarizing micro-environments. It is also
possible to transition from one of the main cell types to another one through the Th0,
TrFoxp3-, Th1R, Th2R or GATA3+IL4- attractors. This ability raises multiple questions about
the signals necessary for plasticity in vivo. It is possible that in order to transition from one cell
type to another, some signals have to be maintained for a certain period of time, or that more
than one perturbation is necessary. Further studies are required to determine which conditions
are necessary and sufficient for CD4+ T cell type transitions to further understand CD4+ T
cell plasticity.
Therefore, in the context of this study, we define plasticity as the potential of a given differ-
entiated cell to attain other fates in response to alterations in the expression patterns of their in-
trinsic components and/or of the extrinsic micro-environment. Of the total of 121 possible
transitions between cell types arising from those alterations, the TSRN network yielded 66 cell-
type transitions. Thus, the topology or set of regulatory interactions proposed in this network
generates restrictions in terms of cell types but also in terms of the patterns of cell-fate
transitions.
CD4+ T cells are typically under the influence of particular micro-environments, with spe-
cific cytokines affecting the dynamics of these cells. Depending on the combination of cyto-
kines, some cell types are lost, and transitions among the remaining cell types are also
restricted. To simulate polarizing micro-environments, we fixed the value of the cytokines as-
sociated with pro-Th1 (IFGγe), pro-Th2 pro-(IL-4e, IL-2e), pro-Th17 (IL-21e, TGF-βe), pro-
iTreg (TGF-βe, IL-2e), pro-Tr (IL-10,) pro-Tfh (IL-21e) and pro-Th9 (IL-4 and TGF-βe). In
general, the polarizing micro-environment increases the size of the attraction basin, the stabili-
ty and the transition into the attractor. The biological nature of the polarizing signal affects the
nature of the resulting transition. In response to regulatory signals (IL-10e, TGF-βe), the ma-
jority of the transitions are towards TrFoxp3-, while inflammatory signals lead to more transi-
tion signals towards Th1 and Th2. All of these results represent interesting predictions that
could be tested experimentally.
Activation of specific CD4+ T transcriptional-signaling regulatory network nodes in-
duces cell type plasticity while inhibitions induce stability.
The nature of the perturbation
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
10 / 23
Fig 5. Cell fate map in response to the micro-environment and perturbations of the TSRN model. The values of the extrinsic signals of the TSRN were
fixed according to different polarizing micro-environments, and the resulting attractors were transiently perturbed. The nodes represent CD4+ T cell types,
and the node sizes correspond to the size of the basin of attraction. The edges represent transitions between cell types, the width of the edges corresponds
to the number of times the transition occurred in logarithmic scale, and self-loops correspond to perturbations where the network returned to the original cell
type. The following micro-environments were studied: combinations of: (A) all extrinsic cytokines, (B) IFN-γe (Th1), (C)IL-4e and IL-2e (Th2), (D) IL-21e and
TGF-βe (Th17), (E) TGF-βe and IL-2e (iTreg), (F) IL-10e (IL10), (G) IL-21e (Tfh), (H) IL-4e and TGF-βe (Th9), (I) no extrinsic cytokines (Th0).
doi:10.1371/journal.pcbi.1004324.g005
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
11 / 23
is also important for CD4+ T cell plastic responses or stability (Fig 6). If an inactive node is ac-
tivated (0!1), there is a high probability that a transition from one cell type to another is in-
duced. In contrast, if an active node is inactivated (1!0), there is a high probability that the
system remains in the original cell-type. This pattern may be caused by the topology of the net-
work and, in particular, may depend on the functional feedback loops of the system that are al-
tered. The positive feedback loops of a cell type may increase the stability of an attractor and
help to recover a transiently inactive node, thus stabilizing a given differentiated state. We hy-
pothesize that the activation of a previously inactive node may induce more transitions, as this
alteration likely affects the positive and negative functional circuits of the system [60], thus in-
creasing the chances that the system leads to a new attractor. Further simulations should be
used to exhaustively test how specific regulatory circuits react to transient activations and inhi-
bitions. In any case, the analysis presented in this study enables us to postulate that CD4+ T
cells are expected to be able to react to activation signals and environmental alterations but are
stabilized in response to the transient loss of signals. Thus, the proposed model for CD4+ T cell
dynamics implies that these cells are under an unstable equilibrium between cell-fate stability
and plasticity.
Key nodes for CD4+ T transcriptional-signaling regulatory network
plasticity
While all the elements of the TSRN have previously been shown to be necessary for the differ-
entiation of CD4+ T cells, we wished to address their relative importance in cell plasticity re-
sponses. To evaluate this question, we perturbed each node of all the attractors and measured
how many times the perturbed state changed to a new attractor (Fig 7) and to which new cell
type the system converged (S4 Fig). This process is equivalent to the temporal activation or in-
activation of a transcription factor or an element of the signaling pathway in response to noise.
Alterations of T-bet and TGF-β usually caused the perturbed state to change from one attractor
to another, while RORγt and IL-9 had the least effect on cell-fate transitions. In general, the
system is more sensitive to perturbations in the master transcriptional regulators than to alter-
ations of the cytokines.
Fig 6. Cell fate map in response to activating or inhibitory signals of the TSRN model. The attractors of
the network were transiently perturbed in all possible micro-environments. Perturbations were considered
activations (0 ! 1) when a previously inactive element was turned on, and inhibitions (1 ! 0) when a
previously active element was turned off. The nodes represent CD4+ T cell types, and the node sizes
correspond to the size of the basin of attraction. The edges represent transitions between cell types, the width
of the edges correspond to the number of times the transition occurred on the logarithmic scale. The number
of transitions towards a different or the original cell type were counted for both activations and inactivations.
doi:10.1371/journal.pcbi.1004324.g006
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
12 / 23
In contrast to previously published T CD4+ network models that only included SOCS1
[37–40], several SOCS-type proteins were considered in the TSRN presented and analyzed
here. SOCS proteins are important for the differentiation and plasticity of CD4+ T cells.
SOCS1 is commonly silenced in inflammatory diseases, and over-expression of SOCS3 corre-
lates with allergies [31,51]. To explore the role of SOCS proteins and the impact of alterations
in these proteins on CD4+ T type transitions, we generated a network lacking the inhibitions
mediated by these proteins (Fig 8). This altered system recovers the original attractors includ-
ing Th0, Th1, Th2, Th17, iTreg, Tfh, TrFoxp3-, and Th9, but it also predicts novel attractors
expressing RORγt+IL-10+ (Th17R) and GATA3+IL-10+IL-9+ (Th2RIL9+), thus confirming
the importance of SOCS proteins for attaining the Th17 and Th9 cell types. The importance of
IL-10 for CD4+ T cell plasticity dramatically increased in the altered network, while the impor-
tance of the rest of the molecular elements decreased. This result suggests that SOCS proteins
play an important role in stabilizing effector cell types and regulating the Th0 and TrFoxp3-
cell types. SOCS proteins inhibit signal transduction; IL-10 in particular acts through these pro-
teins to regulate CD4+ T cells. This regulation is important to buffer the effect of extrinsic cyto-
kines in the TSRN network model. When SOCS proteins are absent, the network is more
sensitive to changes in extrinsic cytokines and IL-10. Further analyses of the effects of SOCS
proteins on CD4+ T cells and the possibility of updates to the model based on experimental
work should enable the evaluation of more subtle alterations in and combinations of SOCS
proteins.
Fig 7. Role of different network nodes in the plasticity of the TSRN model. The proportion of transitions
between attractors in response to transient perturbations in the value of each node. On average, 37.76% of
the perturbations result in transitions to another cell type, with 47.12% of perturbations of intrinsic
components resulting in transitions, compared with 24.43% of perturbations of extrinsic cytokines.
doi:10.1371/journal.pcbi.1004324.g007
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
13 / 23
Discussion
This model provides a mechanistic description of the way in which CD4+ T cell types and plas-
ticity emerge from the interactions among the intrinsic and extrinsic components of the im-
mune response. The study formally shows that, as expected, the interactions among master
transcription factors considered in the TSRN are not sufficient to recover the configurations
characteristic of CD4+ T cell types, nor the reported transition patterns. Furthermore, these re-
sults clearly demonstrate the necessity to include the feedback from signaling pathways in re-
sponse to cytokines to recover most of the range of CD4+ T cell types (Th0, Th1, Th2, Th17,
Tfh, Th9, iTreg and T regulatory Foxp3 independent cells) and their transition pathways.
As noted above, CD4+ T cell differentiation does not arise solely from the regulatory action
of the core of the reported "master" transcription factors (TF): T-bet, GATA3, Foxp3, RORγt
and Bcl6. This may be due to the lack of feedforward loops mediated by the transcription fac-
tors RORγt [48] and Bcl6 [49,50]. These results show that the transcriptional regulatory core of
CD4+ T cell differentiation is necessary, but not sufficient for CD4+ T cell differentiation. The
emergence of the different CD4+ T cell types and their transition patterns, requires the feed-
back from cytokine signaling pathways and external cues.
This model provides a formal test for the emergence of different CD4+ T cell types from
feedback or cooperative dynamics among master transcriptional factors, signaling pathway, cy-
tokines produced by the cell and those present in the micro-environment. The proposed model
recovers the observed configurations for the following CD4+ T cell types: Th0, Th1, Th2,
Th17, Tfh, Th9, iTreg and T regulatory Foxp3 independent cells [2,4–7]. The model also yields
Fig 8. Role of SOCS proteins in the differentiation and plasticity of the TSRN model. The interactions mediated by SOCS proteins were removed to
study their role. (A) Cell fate map of CD4+ T cell types when the SOCS protein interactions are removed from the TSRN model. The nodes represent CD4+ T
cell types and the node sizes correspond to the size of the basin of attraction. New attractors corresponding to GATA3+IL9+IL10+ (Th2RTh9) and RORγt+IL-
10+ (Th17R) appeared. The edges represent transitions between cell types, the width of the edges corresponds to the number of times the transition
occurred on logarithmic scale, and self-loops correspond to perturbations where the network returned to the original cell type. (B) Proportion of transitions
between cell types in response to transient perturbations in the value of each node. On average, 21.65% of the perturbations result in transitions to another
cell type, with 17.55% of perturbations of the intrinsic components of the network resulting in transitions, compared with 27.51% of perturbations of
extrinsic cytokines.
doi:10.1371/journal.pcbi.1004324.g008
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
14 / 23
the cell types Tfh, Th9 and T regulatory Foxp3 independent cells that had not been previously
incorporated into such models [37–40].
CD4+ T cell types depend on signals from other cells for their differentiation and mainte-
nance. The cytokines in the micro-environment restrict which cell types and transitions can be
attained. A cytokine micro-environment that promotes a particular cell type increases its at-
traction basin size, stability and increases the number of transitions towards the promoted cell
type. Nonetheless, different CD4+ T cell types can coexist in micro-environments that do not
promote all the present cell types. For example, the presence of pro-regulatory cytokines IL-10
and TGF-β inhibits most effector cells, except for Th17. This finding may explain the presence
of Th17 cells in regulatory micro-environments [59] and provides important insights concern-
ing the relationship between Th17 and iTreg cells and the paradoxical role of TGF-β in inflam-
mation [61]. Thus, the type of modeling framework and analyses presented here may prove to
be useful for efforts to find therapeutic approaches to address chronic inflammation.
The model was also used to analyze the plasticity of CD4+ T cells by systematically testing
how transient perturbations affect the transition patterns among cell types under various
micro-environments. Previous studies focused on cell plasticity in response to different micro-
environments [38] or on the role of specific molecules [40], rather than studying these phe-
nomena as consequences of the global properties of the system. For example, the TSRN faith-
fully captures the polarization of resting CD4+ T cells into Th1, Th2, Th17, iTreg and
Foxp3-independent T cells, but the predicted cell-fate maps lack direct transitions from iTreg
to Th17 and Th17 to Th1 [23–25]. The TSRN model may lack components, interactions or epi-
genetic mechanisms of regulation that are important to enabling such direct transitions [33].
An additional possibility is that signals must be combined during particular lengths of time to
enable some transitions. Further theoretical and experimental research is required to under-
stand the mechanisms underlying CD4+ T cell plasticity. However, the qualitative model pro-
posed here can serve as a framework to incorporate additional details involved in CD4+ T
plasticity.
Our model shows that the activation of specific CD4+ T cell transcriptional-signaling regu-
latory network nodes generally induce cell type plasticity while inhibitions induce stability. The
observed response patterns may be caused by the feedback loops and mutual inhibitions molec-
ular network. These findings are coherent with the fact that the immune system generates a
specific immunological response to particular challenges, maintains this response while the
challenge remains present, and finally downregulates the immune response once the challenge
has passed, thus maintaining homeostasis [3,61].
Our model suggests that T-bet, TGF-β and SOCS proteins are key network components to
recover the observed CD4+ T cell plasticity. Although T-bet is a key transcription factor for
Th1, it also inhibits other transcription factors regulating the differentiation into different cell
types [4]. TGF-β is a critical regulator of the immune response but also plays a key role during
chronic inflammatory responses [61]. SOCS proteins regulate the phosphorylation of STAT
proteins, playing a key role in modulating the signal transduction among different cell types
[31,51]. Determining the key elements enabling cell-type plasticity has possible therapeutic im-
plications, as these findings can help to identify therapeutic targets for modulating the immune
response while predicting and avoiding secondary effects[3,62].
Given the complexity of CD4+ T cell expression patterns and transitions, it remains unclear
whether cytokine expression profiles correspond to lineages or subsets [1–3,22]. The term line-
age implies the stability of the cellular phenotype and that the cell has committed to an expres-
sion pattern and will maintain it in a fairly robust manner, regardless of environmental
alterations. On the other hand, the term subset implies that the cell has a specified expression
pattern but that extrinsic signals are required to maintain that pattern [1,22]. Cell types Th1,
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
15 / 23
Th2, Tfh and TrFoxp3- can be considered lineages, as they exhibit commitment under different
cytokine milieus, even if the extrinsic signals change, although environmental alterations can
still affect their stability. However, Th17, iTreg and Th9 cells, which require TGF-βe, IL-2e or
IL-4e respectively, are potentially subsets. Th17 and iTreg cells also have small basins of attrac-
tion, low stability, and require extrinsic signals, exhibiting a lack of commitment. Th9 has a
larger basin of attraction than Th17 or iTreg, but is less stable and susceptible to environmental
alterations. Based on our analyses, we propose that the degree of dependence on extrinsic sig-
nals and the stability in response to changes in the micro-environment can provide clearer and
more objective criteria to distinguish between CD4+ T cell subsets and lineages.
CD4+ T cell differentiation and plasticity arises from the feedback among multiple levels of
regulation: transcriptional regulation, signaling pathways and the micro-environment. Study-
ing the molecular network as a dynamic system allows us to understand how the interactions
among the components, the topology of the network, and the dynamic functions of the nodes
give rise to the biological behavior. However, further theoretical and experimental research is
required to understand CD4+ T cells. As our understanding of these cells improves, it will be
possible to incorporate more detailed molecular information, such as the effect of relative ex-
pression levels and the characteristic time courses of expression in the system. This will, in
turn, allow us to recover novel cell types and their relationship with other CD4+ T cell types
and other cells of the immune system. The present model can now be extended to incorporate
multiple cells and their population dynamics [39], relationships with other cells of the immune
system, and the formation of specialized niches that result from the dynamic interaction with
the micro-environment. This approach will allow us to differentiate between CD4+ T cell sub-
sets and lineages, to understand the developmental dynamics between the different cell types,
and to propose approaches to immune system reprogramming that can be used in the clinic.
Methods
Logical modeling formalism: Boolean networks
CD4+ T cell differentiation results from interactions among cytokines, signaling pathways and
transcription factors. These interactions were modeled using Boolean networks that enabled us
to integrate the qualitative nature of complex regulatory systems. A Boolean network is com-
posed of nodes that represent the system´s molecular components (i.e., cytokines, signaling
pathways or transcription factors). In a Boolean network, each node represents a component
(gene, protein, phenomenological signal) that can be associated with a discrete variable denot-
ing its current functional level of activity. If the node is functional its value is 1, and if it is not
functional, then its value is 0 (see S1 File). Some nodes required special considerations concern-
ing their activation states in the Boolean model. For example, in the case of GATA3, which is
continuously expressed during T-cell-lineage development and is necessary for lineage com-
mitment and maintenance, GATA3low is set to 0. As GATA3 is upregulated in Th2 differentia-
tion [63], we set GATA3high to 1. Another example concerns STAT proteins, which are
activated when the protein is phosphorylated, forming a dimer that translocates to the nucleus,
where it activates its target genes. In this case, the value for STAT protein activation was only
set to 1 when all the required conditions were met.
The value of a node xi at a time t depends on the value of the input nodes (including itself),
referred to as its regulators. This value can be expressed with a logical function that describes
the behavior of the node through time:
xiðtÞ ¼ ϕιðτ; ξ1; ξ2; ξ2; …; ξι; …; ξνÞ:
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
16 / 23
Weak interactions that are not necessary or sufficient, but only modulate a target factor,
were not included in the input regulators of the truth tables (S1 File). Such is the case for
Foxp3, which positively modulates the expression of IL-2Rα, which can be activated and func-
tional in the absence of Foxp3 [64].
An input is a node that affects the values of the network but is independent of the network.
The state of the network S can be represented by a vector that specifies the value of each node.
The state of the network can be represented by a vector S composed of the values of all the
nodes of the system. The state of the network corresponds to the expression patterns of a cell.
Inference of the regulatory functions.
Boolean functions were defined based on the avail-
able experimental data for the reported interactions among a network of 85 components (S1
Table). A transcription factor regulates another factor if it binds to the regulatory region of the
latter factor and inhibits or activates is transcription. A cytokine is present if it is either secreted
by the cell (intrinsic) or produced by other cells of the immune system (extrinsic). To separate
the effects of the cytokines produced by the immune system from those of the cytokines pro-
duced by the CD4+ T cell, we label extrinsic cytokines as ILe. Receptors are considered to be ac-
tive when the cytokine is stably bound to a receptor, enabling it to transduce a signal. STAT
proteins are considered active when they are phosphorylated and capable of translocating to
the nucleus. The activation of a STAT protein depends on the presence of interleukin, its cor-
rect binding to the receptor, and subsequent phosphorylation. SOCS proteins inhibit the phos-
phorylation of STAT by competing for the phosphorylation site.
Model reduction
To facilitate the analysis of the network and determine which components were necessary and
sufficient to recover observed profiles and their patterns of transition, we reduced the extended
regulatory network consisting of 85 nodes to one with 18 nodes, including 5 transcription fac-
tors, 7 signaling pathways and 6 extrinsic cytokines. To simplify the network, we assumed that
the signal produced by the TCR and its co-factors was constitutive and ignored weak interac-
tions as well as input and output nodes. Considering that the expression level of node xi at time
t is represented by xi(t), the attractors (steady states) that represent different phenotypes are de-
termined by: xi(t+1) = xi(t).
In that case, the mapping becomes a set of coupled Boolean algebraic equations. The explicit
expressions of the attractors are then obtained by performing the algebraic operations accord-
ing to the axioms of Boolean algebra [44]. Self-regulated nodes were not removed. If a node
was removed, then the logical rules of its targets were modified, maintaining the regulatory
logic and indirect regulation. To verify that we did not remove a necessary node, we recovered
the attractors of the network and ensured that the configurations corresponding to the Th0,
Th1, Th2, Th17 and iTreg states could still be attained (see the details of the reduction methods
used in S1 File).
The reduction was verified using the GINsim[45] software. GINsim uses decision diagrams
to iteratively remove regulatory components and updates the components to maintain the indi-
rect effects. This method preserves the dynamic properties of the original model. The simplifi-
cation with GINsim returned a similar network to the one that we obtained with the Boolean
logic reduction method proposed by Villareal et al. ([44];S1 File).
Dynamic analysis
After inferring and simplifying the network, we studied its dynamic behavior. A regulatory net-
work is a dynamic system. The state of a network will change over time depending on the
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
17 / 23
logical functions associated with each node. When the values of a state vector S at t+1 are the
same as those at time t, the system has attained an attractor: S(t) = S(t + n), n 1.
An attractor is interpreted as a stable expression phenotype of a cell, representing a cell
type. All the states that lead to a solution S constitute the basin of attraction of such an at-
tractor. We determined the attractors and basins of attraction of the network using the R li-
brary BoolNet. Attractors were classified depending on the expression of both the master
transcription factors and the main cytokine. Th0 was defined as expressing no transcription
factors or regulatory cytokines. Th1 was defined as Tbet and IFN-γ active [8], Th2 as GATA3
and IL-4 active [8] and GATA3+ (a Th2-like cell type) as GATA3+IL4-[38]. Th17 was identi-
fied by RORγt and STAT3 signaling mediated by IL-6 or IL-21, all of which require the pres-
ence of TGF-βe [9–10]. The iTreg type was defined by Foxp3 and TGF-β, IL-10 or both, all of
which require the presence of IL-2e [16]. Tfh cells were defined by Bcl6 and STAT3 signaling
mediated by IL-21 [12]. Th9 cells express IL-9, requiring the presence of TGF-β and extrinsic
IL-4 [27]. T regulatory Foxp3-independent CD4+ T cells (TrFoxp3-) featured TGF-β, IL-10 or
both, without expressing Foxp3 [52]. Th1 regulatory cells (Th1R) express a regulatory cytokine
and T-bet [46]. Th2 regulatory cells (Th2rR) express a regulatory cytokine and GATA3 [47].
Network validation.
The network was validated by comparing it with reported knock-out
and over-expression profiles. To simulate loss of function mutations (knock-out) and inhibi-
tions of the signaling pathway, we set the value of the corresponding node to 0 throughout the
complete simulation. To simulate over-expression, the value of the node was set to 1.
The functional robustness of the network was characterized by altering the logical functions
of the network. Functional robustness refers to the invariance of the attractors in response to
noise or perturbations [44]. In this case, to verify that the results of the model did not depend
on over-fitting the logical functions, we perturbed the latter and verified the stability of the re-
sulting attractors and their basins. To achieve this, we randomly selected a large number of en-
tries and flipped their values from 0 to 1 or vice versa, one by one (bit flip). The basins and
attractors were obtained for the altered networks and compared the original basins
and attractors.
To further evaluate the robustness of the network to small changes in the values of the
nodes and interaction functions, we approximated the Boolean step functions as continuous
functions [44]. We replaced the logical functions f(xi) with a set of continuous functions that
satisfy Zadeh's rules of fuzzy propositional calculus. Using this approach for each state variable,
we derived a continuous function, wi(q). The latter functions correspond to step-like (differen-
tiable) activation functions. The continuous system can then be described by:
dqi
dt ¼
1
e½−2bðwiðqÞ−wthr
i
Þþ1 18
where wi is the input function for node i, wi
thr is a threshold level, b is the input saturation rate,
and αi is its relaxation rate. In particular, for b >>1, the activation function becomes a Heavi-
side step function.
Plasticity
The attractors of the network correspond to cell types. A multi-stable system can have multiple
attractors and switch between them in response to alterations in the state of the system [65].
To study the plasticity and robustness of the system we transiently perturbed the attractors of
the network and then evaluated the functions until we arrived at an attractor. This methodolo-
gy enabled us to obtain all the transitions between cell types, the specific perturbations that
caused those transitions, and the path from one cell-type to another. We define an attractor as
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
18 / 23
stable when the system remains in the same attractor in the presence of perturbations. The sta-
bility of each attractor in response to changes in the micro-environment and signaling path-
ways was analyzed by characterizing the evolution of the network in response to pulses of
activation or inhibition of specific nodes. To quantify the stability of the attractors of the net-
work, we perturbed the state vector of the solutions for one time step. Then, we counted how
many of the perturbed state vectors stayed in the same attractor to quantify its stability. A sys-
tem is plastic when it can transition from one state to another in response to alterations of the
system. More specifically, the network was said to be plastic when a transition occurred from a
given attractor to another in response to a transient perturbation in the value of one of
its nodes.
Supporting Information
S1 Table. T CD4+ lymphocyte extended regulatory network references.
(XLS)
S2 Table. T CD4+ lymphocyte transcriptional regulatory network model.
(XLS)
S3 Table. T CD4+ lymphocyte transcriptional-signaling regulatory network model.
(XLS)
S1 File. T CD4+ lymphocyte extended regulatory network simplification.
(PDF)
S2 File. T CD4+ lymphocyte transcriptional-signaling continuous regulatory network
model.
(PDF)
S3 File. Transitions in response to transient perturbations in the nodes of the T CD4+ lym-
phocyte transcriptional-signaling regulatory network.
(PDF)
S1 Fig. T CD4+ lymphocyte extended regulatory network.
(EPS)
S2 Fig. Validation of the T CD4+ lymphocyte transcriptional-signaling regulatory network.
(A) To validate the TSRN model, we simulated loss of function or null mutations (KO) and
over-expression experiments and compared the results with the available experimental data.
The values of the nodes were set to “0” for simulations of loss-of-function or knock-out experi-
ments and to “1” for over-expression. The color corresponds to the basin size of each attractor
on the logarithmic scale. '—-' represents attractors that were not attained in the original wild
type (WT) network. The attractors marked with (red) "X" correspond to incorrect predictions.
(B) To verify the construction of the functions and the structural properties of the model, we
performed a robustness analysis altering the update rules. Networks with perturbed functions
of the TSRN were generated to test the robustness of the structural properties of the networks
to noise, mis-measurements and incorrect interpretations of the data. After altering one of the
functions of the network, 1.389% of the possible initial states changed their final attractor (yel-
low), and only 0.219% of the possible initial states arrived at an attractor not present in the
original network (red).
(EPS)
S3 Fig. Effect of the environment on the stability of the T CD4+ lymphocyte transcription-
al-signaling regulatory network. The values of the extrinsic signals of the TSRN were fixed
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
19 / 23
according to different polarizing micro-environments. Each attractor was transiently per-
turbed, and the proportion of transitions that stayed in the same cell type was plotted on a loga-
rithmic scale. The following micro-environments were studied here: combinations of all
extrinsic cytokines, no extrinsic cytokines (Th0), IFN-γe (Th1), IL-4e and IL-2e (Th2), IL-21e
and TGF-βe (Th17), TGF-βe and IL-2e (iTreg), IL-10e (IL10), IL-21e (Tfh), and IL-4e and
TGF-βe (Th9).
(EPS)
S4 Fig. Effect of transient perturbations on the state of the nodes of the T CD4+ lymphocyte
transcriptional-signaling regulatory network. Number of transitions to an attractor in re-
sponse to transient perturbations in the value of each node. The states of the node were per-
turbed during one time step from 1 to 0 (-) or 0 to 1 (+), depending on its state in the
original attractor.
(EPS)
Acknowledgments
This work is presented in partial fulfillment towards Mariana Martínez-Sanchez’ doctoral de-
gree in the program “Doctorado en Ciencias Biomédicas, de la Universidad Nacional Autón-
oma de México”. We acknowledge Diana Romo for her help with many logistical tasks.
Author Contributions
Conceived and designed the experiments: ERAB MEMS. Performed the experiments: MEMS
CV. Analyzed the data: ERAB MEMS LM CV. Contributed reagents/materials/analysis tools:
MEMS. Wrote the paper: MEMS ERAB.
References
1.
Bluestone JA, Mackay CR, O’Shea JJ, Stockinger B. The functional plasticity of T cell subsets. Nature
Reviews. 2009; 9(11):811–816. doi: 10.1038/nri2654 PMID: 19809471
2.
Sallusto F, Monticelli S. The many faces of CD4 T cells: roles in immunity and disease. Seminars in im-
munology. 2013 Nov; 25(4):249–51. doi: 10.1016/j.smim.2013.11.001 PMID: 24262948
3.
Zak DE, Tam VC, Aderem A. Systems-level analysis of innate immunity. Annual review of immunology.
2014 Jan; 32:547–77. doi: 10.1146/annurev-immunol-032713-120254 PMID: 24655298
4.
Zhu J, Yamane H, Paul WEW, WE ZJYHP. Differentiation of effector CD4 T cell populations. Annual re-
view of immunology. 2010 Jan; 28(1):445–89.
5.
Jiang S, Dong C. A complex issue on CD4(+) T-cell subsets. Immunological reviews. 2013 Mar; 252
(1):5–11. doi: 10.1111/imr.12041 PMID: 23405891
6.
Vahedi G, Kanno Y, Sartorelli V, O’Shea JJ. Transcription factors and CD4 T cells seeking identity:
masters, minions, setters and spikers. Immunology. 2013 Jul; 139(3):294–8. doi: 10.1111/imm.12113
PMID: 23586907
7.
Yamane H, Paul WE. Early signaling events that underlie fate decisions of naive CD4(+) T cells toward
distinct T-helper cell subsets. Immunological reviews. 2013 Mar; 252(1):12–23. doi: 10.1111/imr.12032
PMID: 23405892
8.
Mosmann TR Cherwinski H. Two types of murine helper T cell clone. I Definition according to profiles of
lymphokine activities and secreted proteins J Immunol. 1986; 136:2348–2357. PMID: 2419430
9.
Onishi RM, Gaffen SL. Mechanisms of Interleukin-17 and its target interleukin-17 function in disease.
Immunology. 2010 Mar; 129(3):311–321. doi: 10.1111/j.1365-2567.2009.03240.x PMID: 20409152
10.
Kolls JK, Khader SA. The role of Th17 cytokines in primary mucosal immunity. Cytokine & growth factor
reviews. 2010 Dec; 21(6):443–448.
11.
Breitfeld D, Ohl L, Kremmer E, Ellwart J, et al. Follicular B helper T cells express CXC chemokine re-
ceptor 5, localize to B cell follicles, and support immunoglobulin production. The Journal of experimen-
tal medicine. 2000 Dec; 192(11):1545–52. PMID: 11104797
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
20 / 23
12.
Choi YS, Yang Ja, Crotty S. Dynamic regulation of Bcl6 in follicular helper CD4 T (Tfh) cells. Current
opinion in immunology. 2013 Jun; 25(3):366–72. doi: 10.1016/j.coi.2013.04.003 PMID: 23688737
13.
Putheti P, Awasthi A, Popoola J, Gao W, Strom TB. Human CD4 memory T cells can become CD4+IL-
9+ T cells. PloS one. 2010 Jan; 5(1):e8706. doi: 10.1371/journal.pone.0008706 PMID: 20090929
14.
Lu Y, Hong S, Li H, Park J, Hong B, Wang L, et al. Th9 cells promote antitumor immune responses in
vivo. The Journal of clinical investigation. 2012 Nov; 122(11):4160–71. doi: 10.1172/JCI65459 PMID:
23064366
15.
Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic Self-Tolerance Maintained by Acti-
vated T cells Expressing IL-2 Receptor alpha-Chains (CD25). The Journal of Immunology. 1995 Aug;
155(3):1151. PMID: 7636184
16.
Rudensky AY. Regulatory T cells and Foxp3. Immunological reviews. 2011 May; 241(1):260–268. doi:
10.1111/j.1600-065X.2011.01018.x PMID: 21488902
17.
Rutz S, Ouyang W. Regulation of interleukin-10 and interleukin-22 expression in T helper cells. Current
opinion in immunology. 2011 Oct; 23(5):605–12. doi: 10.1016/j.coi.2011.07.018 PMID: 21862302
18.
Goswami R, Kaplan MH. A brief history of IL-9. Journal of immunology (Baltimore, Md: 1950). 2011
Mar; 186(6):3283–8. doi: 10.4049/jimmunol.1003049 PMID: 21368237
19.
Li H, Rostami A. IL-9: basic biology, signaling pathways in CD4+ T cells and implications for autoimmu-
nity. Journal of neuroimmune pharmacology: the official journal of the Society on NeuroImmune Phar-
macology. 2010 Jun; 5(2):198–209. doi: 10.1007/s11481-009-9186-y PMID: 20020328
20.
Shin K, Wang YH, Santori FR, Boggiano C, Arima K, Bover L, et al. Identification of IL-17-producing
FOXP3+ regulatory T cells in humans. Proceedings of the National Academy of Sciences of the United
States of America. 2009 Mar; 106(12):4793–8. doi: 10.1073/pnas.0900408106 PMID: 19273860
21.
Murphy KM, Stockinger B, Manuscript A. Effector T cell plasticity: flexibility in the face of changing cir-
cumstances. Nature immunology. 2010 Aug; 11(8):674–680. doi: 10.1038/ni.1899 PMID: 20644573
22.
O’Shea J, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells.
Science (New York, NY). 2010 Feb; 327(5969):1098–102. doi: 10.1126/science.1178334 PMID:
20185720
23.
Nakayamada S, Takahashi H, Kanno Y, O’Shea JJ. Helper T cell diversity and plasticity. Current Opin-
ion in Immunology. 2012 Jun; 24(3):297–302. doi: 10.1016/j.coi.2012.01.014 PMID: 22341735
24.
Muranski P, Restifo NP. Essentials of Th17 cell commitment and plasticity. Blood. 2013 Mar; 121
(13):2402–14. doi: 10.1182/blood-2012-09-378653 PMID: 23325835
25.
Hirota K, Duarte JaH, Veldhoen M, Hornsby E, Li Y, Cua DJ, et al. Fate mapping of IL-17-producing T
cells in inflammatory responses. Nature immunology. 2011 Mar; 12(3):255–63. doi: 10.1038/ni.1993
PMID: 21278737
26.
Kleinewietfeld M, Hafler DA. The plasticity of human Treg and Th17 cells and its role in autoimmunity.
Seminars in immunology. 2013 Nov; 25(4):305–12. doi: 10.1016/j.smim.2013.10.009 PMID: 24211039
27.
Kaplan MH. Th9 cells: differentiation and disease. Immunological reviews. 2013 Mar; 252(1):104–15.
doi: 10.1111/imr.12028 PMID: 23405898
28.
Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, Ma L, et al. Generation of T follicular helper cells
is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. Immunity. 2008 Jul;
29(1):138–149. doi: 10.1016/j.immuni.2008.05.009 PMID: 18599325
29.
Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu BP, et al. Molecular antagonism and
plasticity of regulatory and inflammatory T cell programs. Immunity. 2008 Jul; 29(1):44–56. doi: 10.
1016/j.immuni.2008.05.007 PMID: 18585065
30.
Zaretsky AG, Taylor JJ, King IL, Marshall FA, Mohrs M, Pearce EJ, et al. T follicular helper cells differ-
entiate from Th2 cells in response to helminth antigens. The Journal of experimental medicine. 2009
May; 206(5):991–999. doi: 10.1084/jem.20090303 PMID: 19380637
31.
Yoshimura A, Suzuki M, Sakaguchi R, Hanada T, Yasukawa H. SOCS, Inflammation, and Autoimmuni-
ty. Frontiers in Immunology. 2012; 3(March):1–9. doi: 10.3389/fimmu.2012.00001 PMID: 22679445
32.
Cobbold SP, Adams E, Nolan KF, Regateiro FS, Waldmann H. Connecting the mechanisms of T-cell
regulation: dendritic cells as the missing link. Immunological reviews. 2010 Jul; 236:203–18. doi: 10.
1111/j.1600-065X.2010.00913.x PMID: 20636819
33.
Kanno Y, Vahedi G, Hirahara K, Singleton K, O’Shea JJ, Shea JJO. Transcriptional and epigenetic con-
trol of T helper cell specification: molecular mechanisms underlying commitment and plasticity. Annual
review of immunology. 2012 Jan; 30:707–731. doi: 10.1146/annurev-immunol-020711-075058 PMID:
22224760
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
21 / 23
34.
Albert R, Thakar J. Boolean modeling: a logic-based dynamic approach for understanding signaling
and regulatory networks and for making useful predictions. Wiley Interdisciplinary Reviews: Systems
Biology and Medicine. 2014 Sep; 6(5):353–369. PMID: 25269159
35.
Wang RS, Saadatpour A, Albert R. Boolean modeling in systems biology: an overview of methodology
and applications. Physical biology. 2012 Oct; 9(5):055001. doi: 10.1088/1478-3975/9/5/055001 PMID:
23011283
36.
Azpeitia E, Benitez M, Padilla-Longoria P, Espinosa-Soto C, AlvarezBuylla ER. Dynamic network-
based epistasis analysis: boolean examples. Frontiers in plant science. 2011 Jan; 2:92. doi: 10.3389/
fpls.2011.00092 PMID: 22645556
37.
Mendoza L, Pardo F. A robust model to describe the differentiation of T-helper cells. Theory in biosci-
ences Theorie in den Biowissenschaften. 2010 Oct;p. 283–293.
38.
Naldi A, Carneiro J, Chaouiya C, Thieffry D. Diversity and Plasticity of Th Cell Types Predicted from
Regulatory Network Modelling. PLoS Computational Biology. 2010 Sep; 6(9):e1000912. doi: 10.1371/
journal.pcbi.1000912 PMID: 20824124
39.
Mendoza L. A Virtual Culture of CD4+ T Lymphocytes. Bulletin of mathematical biology. 2013 Jan
40.
Carbo A, Hontecillas R, Kronsteiner B, Viladomiu M, Pedragosa M, Lu P, et al. Systems modeling of
molecular mechanisms controlling cytokine-driven CD4+ T cell differentiation and phenotype plasticity.
PLoS computational biology. 2013 Apr; 9(4):e1003027. doi: 10.1371/journal.pcbi.1003027 PMID:
23592971
41.
Chaves M, Albert R. Studying the effect of cell division on expression patterns of the segment polarity
genes. Journal of the Royal Society, Interface / the Royal Society. 2008 Aug; 5 Suppl 1:S71–84. doi:
10.1098/rsif.2007.1345.focus PMID: 18434279
42.
Mendoza L, Sa Ye, Sanchez-Corrales YE, Alvarez-Buylla ER. The Arabidopsis thaliana flower organ
specification gene regulatory network determines a robust differentiation process. Journal of theoretical
biology. 2010 Jun; 264(3):971–83. doi: 10.1016/j.jtbi.2010.03.006 PMID: 20303988
43.
Felix MA. Caenorhabditis elegans vulval cell fate patterning. Physical biology. 2012 Aug; 9(4):045001.
doi: 10.1088/1478-3975/9/4/045001 PMID: 22871570
44.
Villarreal C, Padilla-Longoria P, Alvarez-Buylla ER. General Theory of Genotype to Phenotype Map-
ping: Derivation of Epigenetic Landscapes from N-Node Complex Gene Regulatory Networks. Physical
Review Letters. 2012 Sep; 118102(September):1–5.
45.
Naldi A, Berenguier D, Fauré A, Lopez F, Thieffry D, Chaouiya C. Logical modelling of regulatory net-
works with GINsim 2.3. Bio Systems. 2009 Aug; 97(2):134–9. doi: 10.1016/j.biosystems.2009.04.008
PMID: 19426782
46.
Koch Ma, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ, et al. The transcription
factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nature im-
munology. 2009 Jun; 10(6):595–602. doi: 10.1038/ni.1731 PMID: 19412181
47.
Wohlfert EA, Grainger JR, Bouladoux N, Konkel JE, Oldenhove G, Ribeiro CH, et al. GATA3 controls
Foxp3+ regulatory T cell fate during inflammation in mice. The Journal of clinical investigation. 2011
Nov; 121(11):4503–4515. doi: 10.1172/JCI57456 PMID: 21965331
48.
Ciofani M, Madar A, Galan C, Sellars M, Mace K, Pauli F, et al. A Validated Regulatory Network for
Th17 Cell Specification. Cell. 2012 Sep; 151(2):1–15.
49.
Zhang N, Bevan MJ. Transforming Growth Factor-β Signaling Controls the Formation and Maintenance
of Gut-Resident Memory T Cells by Regulating Migration and Retention. Immunity. 2013 Oct; 39
(4):687–696. doi: 10.1016/j.immuni.2013.08.019 PMID: 24076049
50.
Pasqualucci L, Migliazza A, Basso K, Houldsworth J, Chaganti RSK, Dalla-Favera R. Mutations of the
BCL6 protooncogene disrupt its negative autoregulation in diffuse large Bcell lymphoma. Blood. 2003
Apr; 101(8):2914–23. PMID: 12515714
51.
Ca Knosp, Ja Johnston. Regulation of CD4+ T-cell polarization by suppressor of cytokine signalling
proteins. Immunology. 2012 Feb; 135(2):101–11. doi: 10.1111/j.1365-2567.2011.03520.x PMID:
22044277
52.
Weiner HL. The mucosal milieu creates tolerogenic dendritic cells and T(R)1 and T(H)3 regulatory
cells. Nature immunology. 2001 Aug; 2(8):671–2. PMID: 11477400
53.
Duhen T, Geiger R, Jarrossay D, Lanzavecchia A, Sallusto F. Production of interleukin 22 but not inter-
leukin 17 by a subset of human skin-homing memory T cells. Nature immunology. 2009 Aug; 10
(8):857–863. doi: 10.1038/ni.1767 PMID: 19578369
54.
Zhu J Cote-Sierra J GLPWE, Zhu J, Cote-Sierra J, Guo L, Paul WE, WE ZJCSJGLP. Stat5 activation
plays a critical role in Th2 8differentiation. Immunity. 2003 Nov; 19(5):739–748. PMID: 14614860
55.
et al Kagami S Nakajima H SAHKSK, et al Kagami S Nakajima H SAHKSK. Stat5a regulates T helper
cell differentiation by several distinct mechanisms. Blood. 2001; 97:2358–2365. PMID: 11290598
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
22 / 23
56.
Manel N, Unutmaz D, Littman DR. The differentiation of human T(H)-17 cells requires transforming
growth factor-beta and induction of the nuclear receptor RORgammat. Nature immunology. 2008 Jun;
9(6):641–649. doi: 10.1038/ni.1610 PMID: 18454151
57.
Carrier Y, Yuan J, Kuchroo VKVKK, Weiner HLHLHL. Th3 Cells in Peripheral Tolerance. I. Induction of
Foxp3-Positive Regulatory T Cells by Th3 Cells Derived from TGF-β T Cell-Transgenic Mice. The Jour-
nal of Immunology. 2011; 178(1):179.
58.
Barnes MJ, Powrie F. Regulatory T cells reinforce intestinal homeostasis. Immunity. 2009 Sep; 31
(3):401–11. doi: 10.1016/j.immuni.2009.08.011 PMID: 19766083
59.
Littman DR, Rudensky AY. Th17 and regulatory T cells in mediating and restraining inflammation. Cell.
2010 Mar; 140(6):845–858. doi: 10.1016/j.cell.2010.02.021 PMID: 20303875
60.
Mendoza L, Thieffry D, Alvarez-Buylla ER. Genetic control of flower morphogenesis in Arabidopsis
thaliana: a logical analysis. Bioinformatics. 1999 Jul; 15(7):593–606. PMID: 10341422
61.
McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, et al. TGF-beta
and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology.
Nature immunology. 2007 Dec; 8(12):1390–7. PMID: 17994024
62.
Germain RN, Meier-Schellersheim M, Nita-Lazar A, Fraser IDC. Systems biology in immunology: a
computational modeling perspective. Annual review of immunology. 2011 Apr; 29:527–85. doi: 10.
1146/annurev-immunol-030409-101317 PMID: 21219182
63.
Ho IC, Tai TS, Pai SY. GATA3 and the T cell lineage: essential functions before and after T-helper-2-
cell differentiation. Nat Rev Immunol. 2009 Feb; 9(2):125–135. doi: 10.1038/nri2476 PMID: 19151747
64.
Liao W, Lin JX, Wang L, Li P, Leonard WJ. Modulation of cytokine receptors by IL-2 broadly regulates
differentiation into helper T cell lineages. Nature immunology. 2011 Jun; 12(6):551–559. doi: 10.1038/
ni.2030 PMID: 21516110
65.
van den Ham HJ, de Boer RJ. Cell division curtails helper phenotype plasticity and expedites helper T-
cell differentiation. Immunology and cell biology. 2012 Oct; 90(9):860–8. doi: 10.1038/icb.2012.23
PMID: 22565392
CD4+ T Lymphocyte Minimal Regulatory Network
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004324
June 19, 2015
23 / 23
|
26090929
|
TGFB = ( ( TGFB ) AND NOT ( IL21 ) ) OR ( ( FOXP3 ) AND NOT ( IL21 ) ) OR ( TGFBe )
IL4 = ( ( ( IL4e ) AND NOT ( IL21 ) ) AND NOT ( IFNG ) ) OR ( ( ( ( GATA3 AND ( ( ( IL4 OR IL2 ) ) ) ) AND NOT ( IL21 ) ) AND NOT ( IFNG ) ) AND NOT ( TBET ) )
IL2 = ( ( ( ( IL2e ) AND NOT ( IL21 ) ) AND NOT ( IL10 ) ) AND NOT ( IFNG ) ) OR ( ( ( ( ( IL2 ) AND NOT ( IL21 ) ) AND NOT ( IL10 ) ) AND NOT ( FOXP3 ) ) AND NOT ( IFNG ) )
FOXP3 = ( ( ( IL2 AND ( ( ( FOXP3 OR TGFB ) ) ) ) AND NOT ( RORGT ) ) AND NOT ( IL21 ) )
IL9 = ( ( ( IL4 AND ( ( ( IL10 AND IL2 ) ) OR ( ( TGFB ) ) ) ) AND NOT ( IFNG ) ) AND NOT ( IL21 ) )
RORGT = ( ( ( ( ( IL21 AND ( ( ( TGFB ) ) ) ) AND NOT ( TBET ) ) AND NOT ( BCL6 ) ) AND NOT ( GATA3 ) ) AND NOT ( FOXP3 ) )
IL10 = ( IL10 AND ( ( ( IFNG OR GATA3 OR IL21 OR TGFB ) ) ) ) OR ( IL10e )
BCL6 = ( ( ( ( IL21 ) AND NOT ( IL2 ) ) AND NOT ( TGFB ) ) AND NOT ( TBET ) )
TBET = ( ( ( ( ( ( TBET ) AND NOT ( IL21 ) ) AND NOT ( IL4 ) ) AND NOT ( BCL6 ) ) AND NOT ( IL9 ) ) AND NOT ( GATA3 ) ) OR ( ( ( ( ( ( IFNG ) AND NOT ( IL21 ) ) AND NOT ( IL4 ) ) AND NOT ( BCL6 ) ) AND NOT ( IL9 ) ) AND NOT ( GATA3 ) )
IL21 = ( ( ( ( ( ( IL21 ) AND NOT ( IFNG ) ) AND NOT ( IL9 ) ) AND NOT ( IL2 ) ) AND NOT ( IL10 ) ) AND NOT ( IL4 ) ) OR ( ( ( ( ( ( BCL6 ) AND NOT ( IFNG ) ) AND NOT ( IL9 ) ) AND NOT ( IL2 ) ) AND NOT ( IL10 ) ) AND NOT ( IL4 ) ) OR ( ( ( ( ( ( RORGT ) AND NOT ( IFNG ) ) AND NOT ( IL9 ) ) AND NOT ( IL2 ) ) AND NOT ( IL10 ) ) AND NOT ( IL4 ) ) OR ( ( ( ( ( ( IL21e ) AND NOT ( IFNG ) ) AND NOT ( IL9 ) ) AND NOT ( IL2 ) ) AND NOT ( IL10 ) ) AND NOT ( IL4 ) )
GATA3 = ( ( ( ( ( ( IL4 AND ( ( ( IL2 ) ) ) ) AND NOT ( IL21 ) ) AND NOT ( TBET ) ) AND NOT ( BCL6 ) ) AND NOT ( TGFB ) ) AND NOT ( IFNG ) ) OR ( ( ( ( ( ( GATA3 ) AND NOT ( IL21 ) ) AND NOT ( TBET ) ) AND NOT ( BCL6 ) ) AND NOT ( TGFB ) ) AND NOT ( IFNG ) )
IFNG = ( ( ( ( ( ( ( ( IFNG ) AND NOT ( IL9 ) ) AND NOT ( IL4 ) ) AND NOT ( BCL6 ) ) AND NOT ( IL10 ) ) AND NOT ( TGFB ) ) AND NOT ( GATA3 ) ) AND NOT ( IL21 ) ) OR ( ( ( ( ( ( ( ( TBET ) AND NOT ( IL9 ) ) AND NOT ( IL4 ) ) AND NOT ( BCL6 ) ) AND NOT ( IL10 ) ) AND NOT ( TGFB ) ) AND NOT ( GATA3 ) ) AND NOT ( IL21 ) ) OR ( ( ( ( ( IFNGe ) AND NOT ( IL9 ) ) AND NOT ( IL4 ) ) AND NOT ( IL10 ) ) AND NOT ( IL21 ) )
|
RESEARCH ARTICLE
Inference of Network Dynamics and
Metabolic Interactions in the Gut
Microbiome
Steven N. Steinway1,2☯, Matthew B. Biggs3☯, Thomas P. Loughran Jr2, Jason A. Papin3*,
Reka Albert4*
1 College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States of America,
2 University of Virginia Cancer Center, University of Virginia, Charlottesville, Virginia, United States of
America, 3 Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United
States of America, 4 Department of Physics, Pennsylvania State University, University Park, Pennsylvania,
United States of America
☯These authors contributed equally to this work.
* papin@virginia.edu (JAP); ralbert@phys.psu.edu (RA)
Abstract
We present a novel methodology to construct a Boolean dynamic model from time series
metagenomic information and integrate this modeling with genome-scale metabolic network
reconstructions to identify metabolic underpinnings for microbial interactions. We apply this
in the context of a critical health issue: clindamycin antibiotic treatment and opportunistic
Clostridium difficile infection. Our model recapitulates known dynamics of clindamycin anti-
biotic treatment and C. difficile infection and predicts therapeutic probiotic interventions to
suppress C. difficile infection. Genome-scale metabolic network reconstructions reveal met-
abolic differences between community members and are used to explore the role of metab-
olism in the observed microbial interactions. In vitro experimental data validate a key result
of our computational model, that B. intestinihominis can in fact slow C. difficile growth.
Author Summary
The community of bacteria that live in our intestines (called the “gut microbiome”) is im-
portant to normal intestinal function, and destruction of this community has a causative
role in diseases including obesity, diabetes, and even neurological disorders. Clostridum
difficile is an opportunistic pathogenic bacterium that causes potentially life-threatening
intestinal inflammation and diarrhea and frequently occurs after antibiotic treatment,
which wipes out the normal intestinal bacterial community. We use a mathematical model
to identify how the normal bacterial community interacts and how this community
changes with antibiotic treatment and C. difficile infection. We use this model to identify
bacteria that may inhibit C. difficile growth. Our model and subsequent experiments indi-
cate that Barnesiella intestinihominis inhibits C. difficile growth. This result suggests that
B. intestinihominis could potentially be used as a probiotic to treat or prevent C. difficile
infection.
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
1 / 25
OPEN ACCESS
Citation: Steinway SN, Biggs MB, Loughran TP Jr,
Papin JA, Albert R (2015) Inference of Network
Dynamics and Metabolic Interactions in the Gut
Microbiome. PLoS Comput Biol 11(6): e1004338.
doi:10.1371/journal.pcbi.1004338
Editor: Costas D. Maranas, The Pennsylvania State
University, UNITED STATES
Received: February 18, 2015
Accepted: May 13, 2015
Published: June 23, 2015
Copyright: © 2015 Steinway et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: The code generated
for this paper is publicly available in this repository:
https://bitbucket.org/gutmicrobiomepaper/
microbiomenetworkmodelpaper/src.
Funding: SNS and MBB were funded by The
Jefferson Trust/ University of Virginia Data Science
Institute Collaborative Research Award in Big Data:
http://gradstudies.virginia.edu/bigdata. MBB and JAP
were funded by R01 GM108501 National Institute of
General Medical Sciences, National Institutes of
Health: http://www.nigms.nih.gov/Research/
Mechanisms/Pages/ResearchProjectGrants.aspx.
MBB was funded by University of Virginia
Introduction
Human health is inseparably connected to the billions of microbes that live in and on us. Cur-
rent research shows that our associations with microbes are, more often than not, essential for
our health [1]. The microbes that live in and on us (collectively our “microbiome”) help us to
digest our food, train our immune systems, and protect us from pathogens [2,3]. The gut
microbiome is an enormous community, consisting of hundreds of species and trillions of indi-
vidual interacting bacteria [4]. Microbial community composition often persists for years with-
out significant change [5].
When change comes, however, it can have unpredictable and sometimes fatal consequences.
Acute and recurring infections by Clostridium difficile have been strongly linked to changes in
gut microbiota [6]. The generally accepted paradigm is that antibiotic treatment (or some
other perturbation) significantly disrupts the microbial community structure in the gut, which
creates a void that C. difficile will subsequently fill [7–10]. Such infections occur in roughly
600,000 people in the United States each year (this number is on the rise), with an associated
mortality rate of 2.3% [11]. Each year, healthcare costs associated with C. difficile infection are
in excess of $3.2 billion [11]. An altered gut flora has further been identified as a causal factor
in obesity, diabetes, some cancers and behavioral disorders [12-17].
What promotes the stability of a microbial community, or causes its collapse, is poorly
understood. Until we know what promotes stability, we cannot design targeted treatments that
prevent microbiome disruption, nor can we rebuild a disrupted microbiome. Studying the sys-
tem level properties and dynamics of a large community is impossible using traditional micro-
biology approaches. However, network science is an emerging field which provides a powerful
framework for the study of complex systems like the gut microbiome [18–23]. Previous efforts
to capture the essential dynamics of the gut have made heavy use of ordinary differential equa-
tion (ODE) models [24,25]. Such models require the estimation of many parameters. With so
many degrees of freedom, it is possible to overfit the underlying data, and it is difficult to scale
up to larger communities [26,27]. Boolean dynamic models, conversely, require far less param-
eterization. Such models capture the essential dynamics of a system, and scale to larger systems.
Boolean models have been successfully applied at the molecular [28,29], cellular [20], and com-
munity levels [30]. Here we present the first Boolean dynamic model constructed from metage-
nomic sequence information and the first application of Boolean modeling to microbial
community analysis.
We analyze the dynamic nature of the gut microbiome, focusing on the effect of clindamy-
cin antibiotic treatment and C. difficile infection on gut microbial community structure. We
generate a microbial interaction network and dynamical model based on time-series data from
metagenome data from a population of mice. We present the results of a dynamic network
analysis, including steady-state conditions, how those steady states are reached and main-
tained, how they relate to the health or disease status of the mice, and how targeted changes in
the network can transition the community from a disease state to a healthy state. Furthermore,
knowing how microbes positively or negatively impact each other—particularly for key
microbes in the community—increases the therapeutic utility of the inferred interaction net-
work. We produced genome-scale metabolic reconstructions of the taxa represented in this
community [31], and probe how metabolism could—and could not—contribute to the mecha-
nistic underpinnings of the observed interactions. We present validating experimental evidence
consistent with our computational results, indicating that a member of the normal gut flora,
Barnesiella, can in fact slow C. difficile growth.
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
2 / 25
Biotechnology Training Grant: http://faculty.virginia.
edu/biotech/Home.html. SNS was funded by F30
DK093234 National Institute of Diabetes and
Digestive and Kidney Diseases, National Institutes of
Health: http://grants.nih.gov/grants/guide/contacts/
parent_F30.html. The funders had no role in study
design, data collection and analysis, decision to
publish, or preparation of the manuscript.
Competing Interests: The authors have declared
that no competing interests exist.
Methods
Data Sources
Buffie et al. reported treating mice with clindamycin and tracking microbial abundance by 16S
sequencing [32]. Mice treated with clindamycin were more susceptible to C. difficile infection
than controls. The collection of 16S sequences corresponding to these experiments was ana-
lyzed by Stein et al. [24]. First, Stein et al. aggregated the data by quantifying microbial abun-
dance at the genus level. Abundances of the ten most abundant genera and an “other” group
were presented as operational taxonomic unit (OTU) counts per sample. We use the aggre-
gated abundances from Stein et al. as the starting point for our modeling pipeline (Fig 1).
This processed dataset consisted of nine samples and three treatment groups (n = 3 replicates
per treatment group). The first treatment group (here called “Healthy”) received spores of C. dif-
ficile at t = 0 days, and was used to determine the susceptibility of the native microbiota to inva-
sion. The second treatment group (here called “clindamycin treated”) received a single dose of
clindamycin at t = -1 days to assess the effect of the antibiotic alone, and the third treatment
group (here called “clindamycin+ C. difficile treated”) received a single dose of clindamycin (at t
= -1 days) and, on the following day, was inoculated with C. difficile spores (S1A Fig). Under the
clindamycin+ C. difficile treatment group conditions, C. difficile could colonize the mice and pro-
duce colitis; however this was not possible under the first two treatment group conditions.
Interpolation of Missing Genus Abundance Information
The gut bacterial genus abundance dataset included some variation in terms of time points in
which genera were sampled. That is, genus abundances were measured between 0 to 23 days;
however, not all samples had measurements at all the time points (S1A Fig). Particularly, the
healthy population only included time points at 0, 2, 6, and 13 days and Sample 1 of clindamycin
+ C. difficile treated population was missing the 9 day time point. Missing abundance values for
these 4 points were estimated using an interpolation approach (S1B Fig). For healthy samples,
the 16 and 23 day time points could not be interpolated as the last experimentally identified time
point for these samples is at 13 days. The assumption of the approximated polynomial for these
samples is that extrapolated data points are linear using the slope of the interpolating curve at
the nearest data point. Because genera abundances are fairly stable across time in this treatment
group (i.e. the slope of most of the genera abundances is approximately zero), extrapolating two
time points was deemed reasonable. A principal component analysis was completed on the inter-
polated data (Fig 2A) and shows that the interpolated time series bacterial genus abundance data
clusters by experimental treatment group in the first two principal components. Furthermore,
the results of the binarization for the healthy population suggest that interpolation did not have
any concerning effects on the 16 and 23 day time points (S2 Fig).
Natural cubic spline interpolation was used to estimate genus abundances at missing time
points in some samples. A cubic spline is constructed of piecewise third order (cubic) polyno-
mials which pass through the known data points and has continuous first and second deriva-
tives across all points in the dataset. Natural cubic spline is a cubic spline that has a second
derivative equal to zero at the end points of the dataset [33]. Natural splines were interpolated
such that all datasets had time points at single day intervals through the 23 day time point
(S1B Fig).
Network Modeling Framework
We use a Boolean framework in which each network node is described by one of two qualita-
tive states: ON or OFF. We chose this framework because of its computational feasibility and
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
3 / 25
capacity to be constructed with minimal and qualitative biological data [34]. The ON (logical
1) state means an above threshold abundance of a bacterial genus whereas the OFF (logical 0)
state means below-threshold genus absence. The putative biological relationships among gen-
era are expressed as mathematical equations using Boolean operators [29,34]. We inferred
putative Boolean regulatory functions for each node, which are able to best capture the trends
in the bacterial abundances. These rules, (edges in the interaction network) can be assigned a
direction, representing information flow, i.e. effect from the source (upstream) node to the tar-
get (downstream) node. Furthermore, edges can be characterized as positive (growth promot-
ing) or negative (growth suppressing). An additional layer of network analysis is the dynamic
Fig 1. Dynamic analysis workflow. Time course genus abundance information was acquired from metagenomic sequencing of mouse gastrointestinal
tracts under varying experimental conditions. Missing time points from experimental data were estimated such that genus abundances existed at the same
time points across all treatment groups. Next, genus abundances were binarized such that Boolean regulatory relationships could be inferred. A dynamic
Boolean model was constructed to explore gut microbial dynamics, therapeutic interventions, and metabolic mediators of bacterial regulatory relationships.
doi:10.1371/journal.pcbi.1004338.g001
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
4 / 25
model, which is used to express the behavior of a system over time by characterizing each node
by a state variable (e.g., abundance) and a function that describes its regulation. Dynamic mod-
els can be categorized as continuous or discrete, according to the type of node state variable
used. Continuous models use a set of differential equations; however, the paucity of known
kinetic details for inter-genus and/or inter-species interactions makes these models difficult to
implement.
Binarization
Genus abundance data was binarized (converted to a presence-absence dataset) to enable infer-
ence of Boolean relationships for modeling applications. We adapted a previously developed
approach called iterative k-means binarization with a clustering depth of 3 (KM3) for this pur-
pose [35]. This approach was employed because binarized data is able to maintain complex
Fig 2. Construction of a network model of the gut microbiome from time course metagenomic genus abundance information. Principal component
analysis coefficients associated with each sample in the metagenomic genus abundance dataset was completed for A) interpolated genus abundances and
B) binarized interpolated genus abundances. ‘*’ = Healthy; ‘^’ = clindamycin treated; ‘#’ = clindamycin+ C. difficile treated. C) Consensus binarization of
genus abundance information. Each heatmap represents the consensus binarization for each treatment group. The horizontal axis represents the day of the
experiment that the sample came from. The vertical axis represents the specific genera being modeled. Each genus was binarized to a 1 (ON; above activity
threshold) or 0 (OFF; below activity threshold). D) Interaction rules were inferred from the binarized data. The interaction rules were simplified for visualization
(compound rules were broken into simple one-to-one edges).
doi:10.1371/journal.pcbi.1004338.g002
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
5 / 25
oscillatory behavior in Boolean models constructed from this data, whereas other binarization
approaches fail to maintain these features [35].
Briefly, this approach uses k-means clustering with a depth of clustering d and an initial
number of clusters k = 2d. In each iteration, data for a specific genus G are clustered into k
unique clusters C1
G,. . .,Ck
G, then for each cluster, Cn
G, all the values are replaced by the mean
value of Cn
G. For the next iteration, the value of d is decreased and clustering is repeated. This
methodology is repeated until d = 1. This approach, with d = 3 (called here as KM3 binariza-
tion) has previously been demonstrated as a superior binarization methodology to other binari-
zation approaches for Boolean model construction because it conserves oscillatory behavior
[35]. These analyses were performed using custom Python code based on a previously written
algorithm [35] and is available in the supplemental materials.
Because KM3 binarization has a stochastic component (the initial grouping of binarization
clusters), we employed KM3 binarization on the entire bacterial genus abundance time series
dataset 1000 times. The average binarization for each sample (S2 Fig) was used to determine
the most probable binarized state of each genus in each sample at each time point (S3 Fig). A
principal component analysis of the most probable binarized genus abundances for each sam-
ple demonstrates that as with the continuous time series abundances (Fig 2A), binarized bacte-
rial genus abundance data cluster by experimental treatment group (Fig 2B). For inference of
Boolean rules from the binarized genus abundances (S3 Fig), the consensus of two of three
samples for each treatment population was used as the binarized state of each genus at each
time point in each sample (Fig 2C).
Inference of Boolean Rules from Time Series Genus Abundance
Information
The Best-fit extension was applied to learn Boolean rules from the binarized time series genus
abundance information [36]. For each variable (genus) Xi in the binarized time series genus
abundance data, Best-fit identifies the set of Boolean rules with k variables (regulators) that
explains the variable’s time pattern with the least error size. The algorithm uses partially defined
Boolean functions pdBf (T, F), where the set of true (T) and false vectors (F) are defined as T =
{X0 2 {0, 1}k: Xi (t + 1) = 1} and F = {X0 2 {0, 1}k: Xi (t + 1) = 0}. Intuitively, the partial Boolean
function summarizes the states of the putative regulators that correspond to a turning ON (T) or
turning OFF (F) of the target variable. The error size ε of pdBf(T,F) is defined as the minimum
number of inconsistencies within X0 that best classifies the T and F values of the dataset. The
Best-Fit extension works by identifying smallest size X0 for Xi. For more detailed information
refer to [36]. In line with this, we considered the most parsimonious representation of the rules
with the smallest ε. If the most parsimonious rule was self-regulation, we also considered rules
with the same ε that included another regulator. If multiple rules fit these criteria for a given Xi,
it implied that they can independently represent the inferred regulatory relationships. In cases
where the alternatives had the same value of (non-zero) ε, we explored combinations (such as
appending them by an OR rule) and used the combination that best described the experimentally
observed final (steady state) outcomes. For example, we combined the two alternative rules for
Blautia with an OR relationship. In the case of Barnesiella, we chained three rules ("Other",
"Lachnospiraceae_other", "Lachnospiraceae") by an OR relationship, and "not Clindamycin" by
an AND relationship to incorporate the loss of Barnesiella in the presence of clindamycin (Fig
2C). This was also done for rules for “Lachnospiraceae”, “Lachnospiraceae_other” and “Other”
and all four nodes attained the same rule. There are six nodes with multiple inferred (alternative)
rules: “Barnesiella”,”Blautia”,”Enterococcus”,”Lachnospiraceae”,”Lachnospiraceae_other”, and”-
Other” had 4, 2, 5, 4, 4, and 4 rules, respectively. The six other nodes had a single inferred rule.
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
6 / 25
The network in Fig 2C represents the union of all of the alternative rules produced by Best-Fit,
or in other words,–it is a super-network of all alternative rules. Any alternative networks would
be a sub-network of what we show. A strongly connected component between the nodes inhib-
ited by clindamycin is a feature of the vast majority of these sub-networks. We used the imple-
mentation of Best-Fit in the R package BoolNet [37].
Dynamic Analysis
Dynamic analysis is performed by applying the inferred Boolean functions in succession until a
steady state is reached. Boolean models and discrete dynamic models in general focus on state
transitions instead of following the system in continuous time. Thus, time is an implicit vari-
able in these models. The network transitions from an initial condition (initial state of the bac-
terial community) until an attractor is reached. An attractor can be a fixed point (steady state)
or a set of states that repeat indefinitely (a complex attractor). The basin of attraction refers to
the set of initial conditions that lead the system to a specific attractor. For the network under
consideration, the complete state space can be traversed by enumerating every possible combi-
nation of node states (212) and applying the inferred Boolean functions (or “update rules”) to
determine paths linking those states. The state transition network describes all possible com-
munity trajectories from initial conditions to steady states, given the observed interactions
between bacteria in the community.
We made use of two update schemes to simulate network dynamics: synchronous (deter-
ministic) and asynchronous (stochastic). Synchronous models are the simplest update method:
all nodes are updated at multiples of a common time step based on the previous state of the sys-
tem. The synchronous model is deterministic in that the sequence of state transitions is definite
for identical initial conditions of a model. In asynchronous models, the nodes are updated indi-
vidually, depending on the timing information, or lack thereof, of individual biological events.
In the general asynchronous model used here, a single node is randomly updated at each time
step [38]. The general asynchronous model is useful when there is heterogeneity in the timing
of network events but when the specific timing is unknown. Due to the heterogeneous mecha-
nisms by which bacteria interact, we made the assumption of time heterogeneity without spe-
cifically known time relationships. Synchronous and asynchronous Boolean models have the
same fixed points, because fixed points are independent of the implementation of time. How-
ever, the basin of attraction of each fixed point (i.e. the initial conditions that lead to each fixed
point) may differ between synchronous and asynchronous models (S2 Table). For identifica-
tion of all of the fixed points in the network (the attractor landscape), the synchronous updat-
ing scheme was used. However, for the perturbation analysis, the asynchronous updating
scheme was used because it more realistically models the possible trajectories in a stochastic
and/or time-heterogeneous system. The simulations of the gut microbiome model were per-
formed using custom Python code built on top of the BooleanNet Python library, which facili-
tates Boolean simulations [39]. Our custom Python code is available in the supplemental
materials.
Perturbation Analysis
To capture the effect of removal (knockout) or addition (probiotic; forced over abundance) of
genera, modification of the states/rules to describe removal or addition states were performed.
These modifications were implemented in BooleanNet by setting the corresponding nodes to
either OFF (removal) or ON (addition) and then removing the corresponding updating rules
for these nodes for the simulations. By examining many such forced perturbations, we can
identify potential therapeutic strategies, many of which may not be obvious or intuitive,
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
7 / 25
particularly as network complexity increases. We used asynchronous update when simulating
the effect of perturbations on the microbial communities. In each case we performed 1000 sim-
ulations and report the percentage of simulations that achieve a certain outcome.
Generating Genus-Level Genome-Scale Metabolic Reconstructions
To generate draft metabolic network reconstructions for each of the ten genera in the paper,
we first obtained genome sequences for representative species by searching the “Genomes”
database of the National Center for Biotechnology Information (NCBI). Complete genomes for
the first ten (or if less than ten, all) species within the appropriate genus were downloaded.
During the process of reconstructing genus-level metabolic reconstructions, some genera were
underrepresented (fewer than 10 species genomes) in the NCBI Genome database, including
Akkermansia, Barnesiella and Coprobacillus (S3 Table). The search result order is based on
record update time, and so it is quasi-random. Genomes were uploaded to the rapid annota-
tions using subsystems technology (RAST) server for annotation [40]. Draft metabolic network
reconstructions were generated by providing the RAST annotations to the Model SEED service
[41]. Metabolic network reconstructions were downloaded in “.xls” format. Genus-level meta-
bolic reconstructions were produced by taking the union of all species-level reconstructions
corresponding to each genus, as has been done previously [42]. The one exception was C. diffi-
cile, which was produced by taking the union of three strain-level reconstructions.
Subsystem Enrichment Analysis
Subsystems were defined as the Kyoto Encyclopedia of Genes and Genomes (KEGG) map with
which each reaction was associated [43,44]. These associations were determined based on
annotations in the Model SEED database [41]. To quantify enrichment, the complete set of
unique reactions from all genus-level reconstructions was pooled, and the subsystem annota-
tions corresponding to those reactions were counted. To determine enrichment for a given sub-
set of the community (either a single genus-level reconstruction, or a set of reconstructions
corresponding to a subnetwork), the subsystem occurrences were counted within the subset.
The probability of a reconstruction containing N total subsystem annotations, with M or more
occurrences of subsystem I, was determined by taking the sum of a hypergeometric probability
distribution function (PDF) from M to the total occurrences of subsystem I in the overall popu-
lation. Enrichment analysis was performed in Matlab [45].
Identifying Seed Sets and Defining Metabolic Competition and
Mutualism Scores
To quantify metabolic interactions, we started by utilizing the seed set detection algorithm
developed by Borenstein et al. [46,47]. The algorithm follows three steps:
1. The genome-scale metabolic network reconstruction is reduced into simple one-to-one
edges, such that for each reaction, each substrate and product pair forms an edge (e.g. A + B
! C would become A ! C and B ! C).
2. The network is divided into strongly connected components, those groups of nodes for
which two paths of opposite directions (e.g. A ! B and B ! A) exist between any two
nodes in the group.
3. Nodes (and strongly connected components with five or fewer nodes) for which there are
exclusively outgoing edges are defined as “inputs” to the model, or seed metabolites.
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
8 / 25
The rationale is that metabolites that feed into the network, but cannot be produced by any
reactions within the network, must be obtained from the environment.
Competition metrics were generated following the process of Levy and Borenstein [46]. For
a given pair of genera, the competition score is defined as:
CompScoreij ¼ jSeedSeti \ SeedSetjj
jSeedSetij
ð1Þ
Here SeedSeti is the set of obligatory input metabolites to the metabolic network reconstruc-
tion for genus i, and |SeedSeti| is the number of metabolites contained in SeedSeti. The competi-
tion score indicates the fractional overlap of inputs that genus i shares with genus j, and so
ranges between zero and one. The higher the score, the more similar the metabolic inputs to
the two networks, making competition more likely.
For a given pair of genera, the mutualism score is defined as:
MutualismScoreij ¼ jSeedSeti\:SeedSetjj
jSeedSetij
ð2Þ
Here ¬SeedSetj is the set of metabolites that can be produced by the metabolic network for
species j (i.e. all non-seed metabolites). The mutualism score indicates the fractional overlap of
inputs that genus i consumes which genus j can potentially provide. The mutualism score
ranges between zero and one. The higher the score, the more potential there is for nutrient
sharing between species. While the score does not measure “mutualism” per se (it cannot nec-
essarily distinguish between other interactions such as commensalism or amenalism [48]), for
simplicity, we will refer to these scores as the competition and mutualism scores.
All metabolic reconstructions, seed sets, competition scores and mutualism scores are avail-
able in the supplemental materials. Seed set generation was performed using custom Matlab
scripts, which are available in the supplement. [45]. Statistical tests were performed in R [49].
Co-culture and Spent Media Experiments
Barnesiella intestinihominis DSM 21032 and Clostridium difficile VPI 10463 were grown anaer-
obically in PRAS chopped meat medium (CMB) (Anaerobe Systems, Morgan Hill, CA) at 37
C. To prepare B. intestinihominis spent medium, B. intestinihominis was grown in CMB until
stationary phase (44 hours). The saturated culture was centrifuged, and the supernatant was fil-
ter sterilized (0.22 μM pore size). Growth curves were obtained by inoculating batch cultures in
96-well plates and gathering optical density measurements (870 nm) using a small plate reader
that fits in the anaerobic chamber [50]. Single cultures were inoculated from overnight liquid
culture to a starting density of 0.01. The co-cultures were started at a 1:1 ratio, for a total start-
ing density of 0.02. Optical density was measured every 2 minutes for 24 hours, and the result-
ing growth curves were analyzed in Matlab [45]. Maximum growth rates were calculated by
fitting a smooth line to each growth curve, and finding the maximum growth rate from among
the instantaneous growth rates over the whole time course: [log(ODt+1)—log(ODt)] / [t+1-t].
The achieved bacterial density—area under the growth curve (AUC)—in a culture was calcu-
lated by integrating over the growth curve in each experiment using the “trapz()” function in
Matlab. It can be thought of as representing the total biomass produced over time. The simply
additive null model was calculated by fitting a Lotka-Volterra model [24] to the single cultures
for both B. intestihominis and C. difficile. The null model of co-culture (assuming zero interac-
tion between species) was simulated by using the parameters from single culture, and summing
the predicted OD870 values.
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
9 / 25
All scripts used to analyze the data are available at https://bitbucket.org/
gutmicrobiomepaper/microbiomenetworkmodelpaper/wiki/Home.
Results
Processing of a Microbial Genus Abundance Dataset for Network
Inference
To capture the dynamics of inter-genus interactions in the intestinal tract we employed a pipe-
line (Fig 1) which translates metagenomic genus abundance information into a dynamic Bool-
ean model. This approach involves three steps: 1) discretization (binarization) of genus
abundances, 2) learning Boolean relationships among genera, and 3) translation of genus asso-
ciations into a Boolean (discrete) dynamic model.
Construction of a Dynamic Network Model from Binarized Time Series
Microbial Genus Abundance Information
Boolean rules (S1 Table) were inferred from the time series binarized genus abundances using
an implementation of the Best-fit extension [36] in the R Boolean network inference package
BoolNet [37](see Methods). A network of 12 nodes and 33 edges was inferred (Fig 2D). The
inferred interaction network has a clustered structure: the cluster (subnetwork) containing the
two Lachnospiraceae nodes and Barnesiella is strongly influenced by clindamycin whereas the
other subnetwork is largely independent of the first, except for the single edge between Barne-
siella and C. difficile (Fig 2D). In fact, Lachnospiraceae nodes, Barnesiella and the group of
“Other” genera form a strongly connected component; that is, every node is reachable from
every other node. Most nodes of the second subnetwork are positively influenced by C. difficile,
with the exception of Coprobacillus, for which no regulation by other nodes was inferred, and
Akkermansia, which is inferred to be regulated only by Coprobacillus. These latter two genera
are transiently present (around day 5) in the clindamycin treatment group, but they do not
appear in the final states of any of the treatment groups (see S1 Fig). This network structure is
consistent with published data in which the dominant Firmicutes (Lachnospiraceae) and Bac-
teroidetes (Barnesiella) are devastated by antibiotic administration [51,52]. Furthermore, the
clustered structure (Fig 2D) supports the established mechanism of C. difficile colitis: loss of
normal gut flora, which normally suppresses opportunistic infection (clindamycin cluster), and
the presence of C. difficile at a minimum inoculum (C. difficile cluster) [10,53]. The network
clusters have a single route of interaction between Barnesiella and C. difficile.
The negative influence of Barnesiella on C. difficile is in agreement with recently published
findings in which Barnesiella was strongly correlated with C. difficile clearance [54]. The role of
Barnesiella as an inhibitor of another pathogen (vancomycin-resistant Enterococci (VRE)) has
been shown in mice [55], which is also visible in the network model as an indirect relationship
between Barnesiella and Enterococcus (Fig 2D). Related species of Bacteroidetes have been
shown to play vital roles in protection from C. difficile infection in mice [56]. Furthermore, the
network structure shows that Lachnospiraceae positively interacts with Barnesiella, leading to
an indirect suppression of C. difficile. Interestingly, the two Lachnospiraceae nodes and the
“Other” node form a strongly connected component, suggesting a similar role in the network,
particularly in promoting growth of Barnesiella, which directly suppresses C. difficile. In sup-
port of this finding, Lachnospiraceae has been shown to protect mice against C. difficile coloni-
zation [52,57]. Therefore, the structure of the network is both a parsimonious representation of
the current data set, and is supported by literature evidence.
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
10 / 25
We applied dynamic analysis using the synchronous updating scheme (see Methods) to
determine all the possible steady states of the microbiome network model. In a 12 node net-
work, there are 212 possible network states. We employed model simulations using the syn-
chronous updating scheme to visit all possible network states and identify all fixed points of
the model. Exploration of the steady states of this network reveals 23 possible fixed point
attractors (S4 Fig). Three of the identified attractors (Fig 3A) are in exact agreement with the
experimentally identified terminal time points of binarized genus abundances (Fig 2C). These
attractors make up a small subset of the entire microbiome network state space (S2 Table).
The attractor landscape can be divided into six groups based on abundance patterns they
share (S4 Fig). Group 1 is made up of a single attractor wherein all genera are absent (OFF). The
second group attractor consists of the experimentally defined healthy state (Attractor 2) and gen-
era in the C. difficile subnetwork which can be abundant (ON) independent of the clindamycin
subnetwork. The third grouping has the clindamycin treated steady state (Attractor 7) and gen-
era in the C. difficile subnetwork that can survive in the presence of the clindamycin. Group 4
contains the clindamycin plus C. difficile steady state (Attractor 12) and its subsets in which one
or both of the source nodes Mollicutes and Enterobacteriaceae are absent. Group 5 contains
attractors in which clindamycin is absent and C. difficile is present. Even if clindamycin is absent,
our model suggests that C. difficile can thrive if Lachnospiraceae and Barnesiella are absent, i.e.
these states represent a clindamycin-independent loss of Lachnospiraceae and Barnesiella. Lastly,
group 6 attractors have both clindamycin and C. difficile as OFF. Blautia and Enterococcus are
always abundant in these attractors. Indeed, because of the mutual activation between Blautia
and Enterococcus they always appear together. Attractors in this group may also include the
abundance (ON state) of the source nodes Mollicutes and Enterobacteriaceae.
Perturbation Analysis
We next explored the perturbation of genera in the gut microbiome network model. We con-
sidered the clinically relevant question of which perturbations might alter the microbiome
steady states produced by clindamycin or clindamycin+C. difficile treatment after clindamycin
treatment was removed. Thus, we considered the clindamycin-treated steady state (Attractor 7
in S3 Fig) and the clindamycin+C. difficile treated steady state (Attractor 12) as initial condi-
tions and assumed that clindamycin treatment was stopped. Our simulations, employing asyn-
chronous update (see Methods), indicate that for both initial conditions, only the state of
clindamycin changes after the treatment is stopped; these steady states become Attractor 1 and
Attractor 19, respectively (S4 Fig). In other words, the steady states remain identical in the
absence of clindamycin. We next explored the effect of addition (overabundance; Fig 3B, left
column) and removal (knockout; Fig 3B, right column) of individual genera, simultaneously
with the stopping of clindamycin treatment, on the model predicted steady states. For the per-
turbation analysis, the model was initialized from the clindamycin treated steady state (Fig 3B,
top row) or the clindamycin+C. difficile steady state (Fig 3B, bottom row). From the clindamy-
cin treated state, addition of Lachnospiraceae or “Other” nodes restores the healthy steady
state; however, no removal restore the healthy steady state (Fig 3B). From the clindamycin+C.
difficile state, addition of Barnesiella, Lachnospiraceae, or “Other” nodes lead to a shift toward
the healthy steady state (suppression of C. difficile).
Generating Genus-Level Metabolic Reconstructions
Species-level reconstructions from the genus Enterobacteriaceae contained the most reactions
on average (1335), while those from Mollicutes contained the least (485) (S3 Table). The Barne-
siella and Enterococcus reconstructions contained the most unique reactions (S4 Table) and,
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
11 / 25
interestingly, also displayed more overlap in reaction content between each other (503 reac-
tions) than was observed between any other pair of reconstructions (S5 Table). Lachnospira-
ceae and Barnesiella had the next highest degree of overlap (424 reactions). Mollicutes and
Coprobacillus had the least degree of overlap (363 reactions) (S5 Table). Note that the meta-
bolic reconstructions produced by the SEED framework are draft quality, and as such, may
lack the predictive power of well-curated metabolic reconstructions.
Subsystem Enrichment Analysis
Enrichment analysis was performed for the 99 unique subsystem annotations that were observed
in the community. 22 subsystems displayed interesting enrichment patterns with respect to the
structure of the interaction network (Fig 4). The subsystems for glycolysis/gluconeogenesis and
nucleotide sugars metabolism are enriched in all taxa, highlighting the fact that all taxa contain
Fig 3. Steady states and node perturbations in the gut microbiome model. A) Heatmap of the three steady states in the gut microbiome model. These
steady states are identical to steady states identified in the three experimental groups. B) The effect of node perturbations represented by four heatmaps. On
the Y-axis of each of the four heatmaps are nodes (genera) in each steady state. On the x-axis of each of the four heatmaps are the steady states found
under normal model conditions (i.e. no node perturbations) and also the specific perturbation of a single network node. The two heatmaps in the left column
of the figure demonstrate the effect of addition (forced overabundance) of individual genera, and the two heatmaps in the right column of the figure
demonstrate the effect of removal (knockout) of individual genera. The top row heatmaps show the effect of node perturbations on the clindamycin treated
group and the bottom row heatmaps show the effect of node perturbations on the clindamycin+ C. difficile treatment group. *Genus abundance of 0 means
present in 0% of asynchronous simulations and is indicated in blue; Genus abundance of 1 means present in all (100%) of asynchronous simulations, shown
in yellow. n = 1000 simulations were applied for all Boolean model simulations.
doi:10.1371/journal.pcbi.1004338.g003
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
12 / 25
relatively full complements of reactions within those subsystems. Interestingly, C. difficile is
highly enriched for reactions in cyanamino acid metabolism compared to all other genera.
Lipopolysaccharide (LPS) biosynthesis and cyanoamino acid metabolism subsystems are differ-
entially enriched between C. difficile and both Barnesiella and Lachnospiraceae. Between Barne-
siella and Enterococcus, Barnesiella is more highly enriched for d-glutamine and d-glutamate
metabolism, pantothenate and CoA biosynthesis, LPS biosynthesis. With respect to Enterococ-
cus, Barnesiella is less highly enriched in pyrimidine metabolism, and phenylalanine, tyrosine,
and tryptophan biosynthesis.
Fig 4. Subsystem enrichment analysis highlights metabolic differences between taxa. The p-values from the enrichment analysis are log-transformed
and negated, such that darker regions indicate greater enrichment. The enrichment analysis quantifies the likelihood that a given subsystem (row) would be
as highly abundant as observed within a given metabolic reconstruction (column) by chance alone. A subset of 22 interesting subsystems is shown here.
Subsystems of interest include those for which all taxa are enriched, such as glycolysis, and nucleotide sugars metabolism, highlighting the fact that all taxa
contain relatively full compliments of reactions within those subsystems. Similarly, subsystems for which a single genus differs from the remaining genera are
interesting, such as cyanoamino acid metabolism, where C. difficile is highly enriched for reactions in that subsystem. Some subsystems are differentially
enriched between Barnesiella and Lachnospiraceae, and C. difficile such as lipopolysaccharide biosynthesis and cyanoamino acid metabolism.
doi:10.1371/journal.pcbi.1004338.g004
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
13 / 25
Generating Metabolic Competition and Mutualism Scores
The metabolic reconstructions were used to explore the potential metabolic underpinnings of
the inferred interaction network. Competition scores were generated for all pairwise relation-
ships between the genera considered in the model (self-edges were excluded). The two Lachnos-
piraceae genera were treated as metabolically identical, and the “Other” group was excluded. We
grouped pairs of genera into five groups based on being connected by a positive or negative edge,
a negative or positive path (meaning an indirect relationship), or no path. A positive relationship
was found between competition score and edge type in the interaction network (i.e. positive
edges tend to have a higher competition score), which was not statistically significant, perhaps
due to the small sample size (p-value = 0.058 by one-sided Wilcoxon rank sum test) (S5A Fig).
The mutualism score did not display any obvious trends with respect to the network structure
(S5B Fig). All pairs with inferred edges exhibited relatively high competition scores and low
mutualism scores (S5C Fig). Barnesiella, a key inhibitor of C. difficile in the interaction network,
holds the second smallest competition score with C. difficile (see Fig 5A). Barnesiella and C. diffi-
cile also have the highest mutualism score among all interacting pairs in the network (S5C Fig).
The positive relationship between edge type and competition score suggests that more meta-
bolic similarity between genera tends to foster positive interaction. The converse is also true,
where less metabolic similarity tends to foster negative interactions (S5A Fig). Here, “positive/
negative interaction” is derived from the Boolean model, where a positive edge between species
A and B indicates that if A is ON at time t, then B is likely to turn ON at t+1.
Co-culture and Spent Media Experiments
Barnesiella intestinihominis was chosen as a representative species for the genus Barnesiella for the
in vitro experiments. C. difficile grew more slowly in B. intestinihominis spent media (n = 16, p-
value < 0.005, by one-sided Wilcoxon rank sum test) (Fig 5B). The co-culture with both B. intesti-
nihominis and C. difficile grew more slowly than C. difficile alone (n = 16, p-value < 0.05, by one-
sided Wilcoxon rank sum test) (Fig 5B). C. difficile area under the growth curve (AUC), a measure
of the achieved bacterial density over the experiment, was not statistically different between growth
in fresh media and B. intestinihominis spent media (n = 16, p-value = 0.22 by one-sided Wilcoxon
rank sum test). However, the co-culture displayed a much lower AUC than expected under a null
model of interaction (in which the two species do not interact) (Fig 5C). Examining the co-culture
growth curve, it maintained a consistently lower density than a null model (Fig 5D).
Discussion
Here we have developed a novel strategy for generating a dynamic model of gut microbiota com-
position by inferring relationships from time series metagenomic data (Fig 1). To our knowledge,
this is the first Boolean dynamic model of a microbial interaction network and the first Boolean
model inferred from metagenomic sequence information. Metagenomic sequencing is a power-
ful tool that tells us the consequences of microbial interaction—changes in bacterial abundance.
Bacterial interactions are, in fact, mediated by the many chemicals and metabolites the bacteria
use and produce. In a network sense these relationships are a bipartite graph; bacterial genera
produce chemicals/metabolites, which have an effect on other bacteria. Because there is no com-
prehensive source for the bacterial metabolites and their effect on other bacterial genera, we infer
the effects of genera on each other from the relative abundances of genera in a set of microbiome
samples, and we employ genome-scale metabolic reconstructions to gain insight into these rela-
tionships (Fig 6B). Binarization of the microbial abundances clarifies these relationships and is
the starting point for the construction of a dynamic network model of the gut microbiome. Inter-
estingly, principal component analysis demonstrates that the time series data clusters by
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
14 / 25
experimental treatment group, suggesting that our initial assumption of binary relationships
does not lead to significant information loss (Fig 2A and 2B).
Fig 5. Metabolic competition scores and in vitro data indicate a non-metabolic interaction mechanism. A) Competition scores for all pairs of genera
with C. difficile. Notice that Barnesiella has nearly the lowest competition score. B) Maximum growth rates for all growth conditions. C. difficile grew more
slowly in B. intestinihominis spent media (n = 16, p-value < 0.005, by one-sided Wilcoxon rank sum test). The co-culture with both B. intestinihominis and C.
difficile grew more slowly than C. difficile alone (n = 16, p-value < 0.05, by one-sided Wilcoxon rank sum test). C) Area under the curve (AUC) was not
significantly different for C. difficile in fresh media or B. intestinihominis spent media (n = 16, p-value = 0.22 by one-sided Wilcoxon rank sum test). D) The
experimental (red, solid line) and simulated (blue, dashed line) co-culture growth curves. “Binte” indicates B. intestinihominis, while “Cdiff” stands for C.
difficile. On average, the experimental co-culture growth curves maintained a lower density than the simply additive null model. Error bars represent the
standard error of the mean from 16 independent replicates.
doi:10.1371/journal.pcbi.1004338.g005
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
15 / 25
We analyze the topological and dynamic nature of the gut microbiome, focusing on the
effect of clindamycin antibiotic and C. difficile infection on gut microbial community structure.
We generate a microbial interaction network and dynamic model based on time-series data
from a population of mice. We validate a key edge in this interaction network between Barne-
siella and C. difficile through an in vitro experiment. Consistent with the literature, our model
affirms that solely inoculating a healthy microbiome with C. difficile is insufficient to disrupt
the healthy intestinal tract microbiome. Additionally, our results demonstrate that clindamycin
treatment has a tremendous effect on the microbiome, greatly reducing many microbial genera,
and that during the time C. difficile is present, a certain subset of bacteria come to dominate the
microbiome (S1 and S2 and 2C Figs).
Our dynamic network model reveals the steady state conditions attainable by this microbial
system, how those steady states are reached and maintained, how they relate to the health or
disease status of the mice, and how targeted changes in the network can transition the
Fig 6. Computational models can bring us closer to true interaction networks. A) Potential inhibitory mechanisms include direct inhibition of C. difficile
by Barnesiella (e.g. via competition for scarce resources, or toxin production), or indirect inhibition (e.g. through a host antimicrobial response). B) A great
deal has been published on the topic of network inference from complex data sets, and more can be done to improve inference methods. Particularly for
microbial interaction networks, it is essential to identify, not only the nature of the interactions, but also the underlying mechanisms. Metagenomic genus
abundance information can be used to infer causal relationships between bacteria; however, other information sources are required to determine the exact
nature of these interactions. Each individual network edge may have very different underlying causes (metabolic, physical interaction, toxin-based, etc.).
Including more tools in the pipeline, such as metabolic network reconstructions, bioinformatics tools, etc., will help elucidate these mechanisms, allowing far
more rapid hypothesis generation, leading to a more focused effort in the wet lab.
doi:10.1371/journal.pcbi.1004338.g006
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
16 / 25
community from a disease state to a healthy state. Furthermore, we examine genome-scale
metabolic network reconstructions of the taxa represented in this community, examine broad
metabolic differences between the taxa in the community, and probe how metabolism could—
and could not—contribute to the mechanistic underpinnings of the observed interactions.
Network Structure
The first feature that stands out in the inferred interaction network is its clustered structure.
Clindamycin has a strong influence on the subnetwork containing the two Lachnospiraceae
nodes and Barnesiella. The other subnetwork contains C. difficile and other genera that become
abundant during C. difficile infection (Fig 2D). Also worth noticing are the two contradicting
edges in the network, between Coprobacillus and Blautia, and the self-edges for Blautia (Fig
2D). These arise from rules in the Boolean model that are context-dependent. Such context-
dependent rules can manifest as opposite edge types, depending on the state of other nodes in
the network. Context-dependent interactions have been demonstrated in many microbial pair-
ings, and nutritional environments can even be designed to induce specific interaction types
[58]. It is possible that subtle environmental changes over the course of the experiment altered
conditions in a way that flipped the Coprobacillus-Blautia interaction. Because the interaction
network is derived from time-series data, it is possible to estimate causality, and therefore,
derive a directed graph. A directed network with clear, causative interactions can be used to
study community dynamics. This is in contrast with association networks, which are often
derived from independent samples, and cannot determine direction of causality [48,59–61].
Such networks are more limited in utility because they cannot be used to predict system behav-
ior over time, or system responses to perturbations [24,62]. Note that the inferred network
structure represents a set of hypotheses as to potential interactions among genera. Determining
whether or not the interactions truly occur requires further experimentation, similar to the
experimentation completed to validate the edge between Barnesiella and C. difficile.
Experimental Validation of Barnesiella Inhibition of C. difficile
We experimentally validated a key edge in the interaction network, and showed that Barnesiella
can in fact slow C. difficile growth. C. difficile was grown alone, in co-culture with B. intestini-
hominis, and in B. intestinihominis spentmedia. C. difficile grew more slowly in both co-culture
and spent-media conditions. Though moderate, the effect was statistically significant (Fig 5B).
The fact that C. difficile growth rate was inhibited under spent-media conditions indicates that
B. intestinihominis-mediated inhibition does not require B. intestinihominis to “sense” the
presence of C. difficile. Further, C. difficile growth on B. intestinihominis spent media demon-
strates that the two species have different nutrient requirements. Whether the reduction in
growth rate is a result of nutritional limitations (e.g. C. difficile resorts to a less preferred carbon
source) is unknown, but unlikely given the AUC data.
The AUC—a summation of the OD over the entire time course—is a measure of the total
bacterial density achieved over the course of the experiment. It can be thought of as a single
metric combining growth rate and biomass production over time. Examining the AUC for all
conditions showed that C. difficile AUC did not significantly change between fresh media and
spent media (Fig 5C). Thus, C. difficile was able to produce comparable overall biomass despite
a reduction in growth rate, further demonstrating that nutrient availability was sufficient in the
spent media condition. The AUC for the co-culture was much lower than expected in a simu-
lated null model (Fig 5C). Apparently, in co-culture, the total community biomass production
capacity is reduced from what would be expected in a scenario without species interaction.
Thus, there is a measurable negative interaction between B. intestinihominis and C. difficile in
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
17 / 25
co-culture that impacts biomass production. This can be observed over the full time-course of
the co-culture, where the overall density is consistently lower than what would be expected in a
null model (Fig 5D).
Network Dynamics and Perturbation Analysis
Computational perturbation analysis showed that forced overabundance of Barnesiella led to a
shift from the “disease” state (clindamycin+ C. difficile treatment group) to a state highly simi-
lar to the original healthy state (loss of C. difficile). This result is particularly interesting from a
therapeutic design standpoint. In this case, the model results indicate that Barnesiella may
serve as an effective probiotic. Model-driven analysis can be used to identify candidate organ-
isms for probiotic treatments. Recent work by Buffie et al. performed a proof-of-concept study
in which they used statistical models to identify candidate probiotic organisms, which were
then tested on a murine model of C. difficile infection [54]. This model-driven approach can be
favorably contrasted with the brute-force experimental approach in which successive combina-
tions of microbes are tested until a curative set is found [56]. The model-driven approach
requires far fewer experiments, and saves time and resources. While the computational model
presented here differs from that used by Buffie et al., the integration of computational models
in probiotic design has been shown to be a feasible, effective approach. Improved tools, such as
the perturbation analysis presented here, will surely accelerate the probiotic design process and
shorten the path to the clinic.
Metabolic Competition Scores Point towards a Non-metabolic
Interaction Mechanism
Genome-scale metabolic network reconstructions can be used to estimate the interactions
between microbes in a complex community based purely on genome sequence data. Our use of
genus-level metabolic network reconstructions (a union of several species-level reconstruc-
tions) may not reflect the unique, species-level interactions and heterogeneity within a commu-
nity. This higher-level model will only capture broad trends and the possible extent of
metabolic capacity within a genus. Furthermore, the draft status of these models precludes the
effective application of flux balance analysis (FBA) to estimate interactions among genera. This
is due to the established lack of precision in draft reconstructions in predictions of growth rates
and substrate utilization patterns [63], and the sensitivity of interaction models to metabolic
environment and model structure [58,64]. Future efforts to infer metabolic interactions using
FBA and well-curated metabolic networks could provide deeper insights into specific metabo-
lites that are shared (or competed for) between specific microbial pairs.
The application of competition scores demonstrated here (S5A Fig) could potentially be
used to quickly establish a rough expectation (notice the spread of competition scores for the
species pairs not connected by a path through the network) for community structure—based
solely on genomic information—that can then be tested experimentally. Interestingly, the fact
that higher competition score is associated with more positive interactions inferred from the
Boolean model relates to previous work that demonstrates that higher competition scores were
associated with habitat co-occurrence [46]. In this same work, the authors suggest that this
effect is due to habitat filtering; that is, microbes with similar metabolic capabilities tend to
thrive in similar environments. It has been shown experimentally that microorganisms from
the same environment tend to lose net productivity in batch co-culture, indicating similar met-
abolic requirements [65]. Thus, it appears that metabolically similar organisms tend to co-
locate to similar niches, and over evolutionary time, co-localized organisms tend to develop
positive relationships with each other.
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
18 / 25
Understanding this relationship between competition score and interaction type leads to the
conclusion that negative interactions are probably not caused by metabolic competition. Of all
the genus competition scores with C. difficile, Barnesiella showed the second lowest (Fig 5A). In
other words, Barnesiella is among the least likely to share a similar metabolic niche with C. diffi-
cile, which fits with the broad trend mentioned above. The fact that the competition score
between C. difficile and Barnesiella is so low strongly suggests that the negative interaction
between them is due, not to competition for scarce resources (although it does not completely
exclude the possibility), but rather to some non-metabolic mechanism. The similarity in reaction
content between Barnesiella and Enterococcus indicates similar network structure (S5 Table),
and yet, Enterococcus does not inhibit C. difficile in the inferred interaction network (Fig 2D).
Either the differences that are present between Barnesiella (65 unique reactions) and Enterococ-
cus (36 unique reactions) are hints at the mechanism of interaction, or metabolism does not play
a significant role in C. difficile inhibition in the environment of the gut. For example, enrichment
analysis showed that that, with respect to Enterococcus, Barnesiella is more highly enriched for
d-glutamine and d-glutamate metabolism, pantothenate and CoA biosynthesis and LPS biosyn-
thesis. With respect to Enterococcus, Barnesiella is less enriched in pyrimidine metabolism, and
phenylalanine, tyrosine, and tryptophan biosynthesis. The possible role of LPS is discussed fur-
ther on. The possible roles of these other metabolic pathways in C. difficile inhibition is unclear.
There is experimental evidence that Barnesiella (and other normal flora) may combat path-
ogen overgrowth through non-metabolic mechanisms. As a first step, it has been shown that
VRE can grow in sterile murine cecal contents—indicating the presence of sufficient nutrition
to support VRE—but is inhibited in saline-treated cecal contents—indicating that live flora are
needed to suppress VRE growth, and that this suppression is not through nutrient sequestra-
tion [66]. Further, the presence of B. intestinihominis has been demonstrated to prevent and
cure VRE infection in mice [55], and is strongly correlated with resistance to C. difficile infec-
tion in mice [54]. Clearly, Barnesiella plays a key role in pathogen inhibition, and pathogen
inhibition can be caused by mechanisms other than nutrient competition.
This non-metabolic mechanism may be direct or indirect (Fig 6A). We demonstrated in
vitro that B. intestinihominis can inhibit C. difficile growth rate (Fig 5C and 5D). The fact that
C. difficile grows on B. intestinihominis spent media at all indicates that the metabolic require-
ments of the two species are different, which is consistent with our computational results sup-
porting the hypothesis that C. difficile and Barnesiella do not compete metabolically (Fig 5B).
Further, C. difficile is moderately inhibited both in co-culture with B. intestinihominis and in B.
intestinihominis-spent media, indicating a direct mechanism of inhibition. In further support
of a direct mechanism, it has been shown that Clostridium scindens inhibits growth of C. diffi-
cile through the production of secondary bile acids [54]. Perhaps Barnesiella works through an
analogous mechanism in vivo, enhancing the moderate inhibition observed in vitro.
In support of an additional indirect mechanism of bacterial interaction, Buffie and Pamer,
in a recent review, hypothesized that the normal flora (of which Barnesiella is a member) may
prevent pathogen overgrowth by stimulation of a host antimicrobial response [67] (Fig 6A).
Specifically, they point out that Barnesiella can activate host toll-like receptor TLR signaling,
which activates host antimicrobial peptide production. For example, LPS and flagellin have
been shown to stimulate the host innate immune response through toll-like receptor (TLR) sig-
naling and production of bactericidal lectins [68,69]. Barnesiella shows enrichment for LPS
biosynthesis pathways (Fig 4). However, this mechanism did not seem to be responsible for
inhibition of VRE by Barnesiella [55]. An indirect, host-mediated mechanism is further sup-
ported by the fact that members of the normal gut flora can interact differently with pathogens
depending on the host organism [54]. Regardless, any indirect mechanism is in addition to the
direct inhibitory mechanism observed in vitro. Both direct and indirect mechanisms may play
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
19 / 25
a role in vivo, and further work is needed to clearly discern the underlying process that allows
Barnesiella to play this protective role.
We demonstrate that dynamic Boolean models capture key microbial interactions and
dynamics from time-series abundance data in a murine microbiome. We show that this
computational approach enables exhaustive in silico perturbation, which leads to fast candidate
selection for probiotic design. We further describe the use of genome-scale metabolic network
reconstructions to explore the metabolic potential attributed to community members, and to
estimate metabolic competition and cooperation between members of the microbiome com-
munity. Analysis of genome-scale metabolic network reconstructions indicates that Barnesiella
likely inhibits C. difficile through some non-metabolic mechanism. We present empirical in
vitro evidence that B. intestinihominis does in fact inhibit C. difficile growth, likely by a non-
metabolic mechanism, and our findings are in good agreement with published results. We
present this work as a demonstration of the use of dynamic Boolean models and genome-scale
metabolic reconstructions to explore the structure, dynamics, and mechanistic underpinnings
of complex microbial communities.
Supporting Information
S1 Fig. Bacterial genera abundances over time in response to clindamycin treatment and/
or C. difficile inoculation. A) Genera abundance information for the nine samples. The
“Healthy” population received spores of C. difficile (at t = 0 days) and did not undergo observ-
able microbial changes, Population 2 received a single dose of clindamycin (at t = -1 days), and
Population 3 received a single dose of clindamycin (at t = -1 days) and, on the following day,
was inoculated with C. difficile spores (at t = 0 days). Genus abundances were measured at 0, 2,
3, 4, 5, 6, 7, 9, 12, 13, 16, and 23 days; however, not all samples had measurements at all the
time points. B) Cubic spline interpolation of data points was performed such that all the same
time point measurements of bacterial abundance occurred in all samples and that single day
intervals were present in all datasets.
(TIF)
S2 Fig. Averaged binarized genera abundances using iterative k-means binarization. Itera-
tive k-means binarization was completed on all the samples 1000 times and average binariza-
tion is shown for each genus at each time point in each of the nine samples. If a node (genus) is
binarized as 0 (OFF) at a time step, then it is colored blue, and if a node (genus) is binarized as
1 (ON) at a time step, then it is colored yellow. This figure represents the average of 1000 repli-
cates of IKM binarization. Intermediate cell colors represent cases where a genus abundance at
a time point was binarized to 1 (ON) in a fraction of the replicates.
(TIFF)
S3 Fig. Averaged binarized genera abundances using iterative k-means binarization were
rounded to the most probable binarized state. The most probable binarized state of each
genus at each time point. If the average genus abundance binarization (S2 Fig) was greater than
0.5 (ON in over 500 of 1000 replicates), then that genus abundance was assumed to be 1 (ON)
for downstream analysis. If the average genus abundance binarization was less than 0.5 (ON in
less than 500 of 1000 replicates) then that genus abundance was assumed to be 0 (OFF) for
downstream analysis.
(TIFF)
S4 Fig. All possible steady states of the Boolean model of the gut microbiome. There are 23
predicted steady states in the Boolean model of the gut microbiome. Each attractor is a column
in the heatmap and is made up of the state of each genus in the network model (rows). Each
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
20 / 25
genus can be present above an activity threshold (yellow; ON) or below an activity threshold
(blue; OFF). The steady states in the model are grouped based on their similarities to other
steady states in the same group. The first steady state of group 2 (Attractor 2) is the healthy
steady state, the first steady state of group 3 (Attractor 7) is the clindamycin treated steady
state, and the first steady state of group 4 (Attractor 12) is the clindamycin + C. difficile steady
state. These three steady states are directly corroborated by experimental metagenomic data.
(TIFF)
S5 Fig. Competition and mutualism scores by edge and path type in Boolean network. A)
Competition score values for all classes of paths through the network, including direct edges,
directed paths, and no directed path. A positive relationship was found between competition
score and direct edge type in the dynamic network (self-edges were excluded), which was not
statistically significant, perhaps due to the small sample size (p-value = 0.058 by one-sided Wil-
coxon rank sum test), but is worthy of note. B) Mutualism score values for all classes of paths
through the network, including direct edges, directed paths, and no directed path. C) Competi-
tion and mutualism score plot for the interaction edges in the network. All the interactions
reflect moderate to high competition scores and relatively low mutualism scores. All the inter-
actions have a higher competition score than mutualism score. The two negative interactions
(red circles) do not have higher competition scores, nor lower mutualism scores, than the posi-
tive interactions. In fact, the negative interaction between Barnesiella and C. difficile corre-
sponds to the highest mutualism score.
(TIF)
S1 Table. Boolean update rules for the gut microbiome network. The ruleset inferred from
metagenomic sequencing information using Boolnet.
(DOCX)
S2 Table. Basin size as % of total state space (unique basin size) for experimentally realized
network attractors.
(DOCX)
S3 Table. Genus-level genome-scale metabolic network reconstruction characteristics. In
this table we characterize the genus-level metabolic network reconstructions. Average model
size refers to the average number of reactions in the component species reconstructions within
each genus. Akkermansia is represented by a single species-level reconstruction, while several
genera are represented by 10 species-level reconstructions. The average network overlap within
a genus refers to the average number of shared reactions between any two pairs of species
within the genus. Similarly, the average fraction of unique reactions refers to the average subset
of reactions in a given species that are unique within the genus.
(DOCX)
S4 Table. Unique reactions within genera. The genus in each row has n reactions that the
genus in the columns do not have. For example, the genus-level reconstruction for Barnesiella
contains 167 reactions that the reconstruction for C. difficile does not. Conversely, the recon-
struction for C. difficile only contains 30 unique reactions that the reconstruction for Blautia
does not already contain.
(DOCX)
S5 Table. Reaction overlap between genera. The upper portion of the table contains the num-
ber of shared reaction content between all genus-level metabolic network reconstructions.
(DOCX)
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
21 / 25
Acknowledgments
The authors thank Dr. Glynis Kolling (University of Virginia) for help obtaining bacterial iso-
lates and carrying out in vitro experiments. The authors further thank Dr. David J. Feith (Uni-
versity of Virginia) for helpful comments/suggestions.
Author Contributions
Conceived and designed the experiments: SNS MBB JAP RA. Performed the experiments: SNS
MBB. Analyzed the data: SNS MBB TPL JAP RA. Contributed reagents/materials/analysis
tools: SNS MBB JAP RA. Wrote the paper: SNS MBB TPL JAP RA.
References
1.
Gordon JI (2012) Honor thy gut symbionts redux. Science 336: 1251–1253. doi: 10.1126/science.
1224686 PMID: 22674326
2.
Bergman EN (1990) Energy contributions of volatile fatty acids from the gastrointestinal tract in various
species. Physiol Rev 70: 567–590. PMID: 2181501
3.
Rosenberg E, Sharon G, Atad I, Zilber-Rosenberg I (2010) The evolution of animals and plants via sym-
biosis with microorganisms. Environ Microbiol Rep 2: 500–506. doi: 10.1111/j.1758-2229.2010.00177.
x PMID: 23766221
4.
Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI (2011) Human nutrition, the gut microbiome
and the immune system. Nature 474: 327–336. doi: 10.1038/nature10213 PMID: 21677749
5.
Faith JJ, Guruge JL, Charbonneau M, Subramanian S, Seedorf H, et al. (2013) The long-term stability
of the human gut microbiota. Science 341: 1237439. doi: 10.1126/science.1237439 PMID: 23828941
6.
Reeves AE, Theriot CM, Bergin IL, Huffnagle GB, Schloss PD, et al. (2011) The interplay between
microbiome dynamics and pathogen dynamics in a murine model of Clostridium difficile Infection. Gut
Microbes 2: 145–158. PMID: 21804357
7.
Bartlett JG, Moon N, Chang TW, Taylor N, Onderdonk AB (1978) Role of Clostridium difficile in antibi-
otic-associated pseudomembranous colitis. Gastroenterology 75: 778–782. PMID: 700321
8.
George WL, Rolfe RD, Finegold SM (1982) Clostridium difficile and its cytotoxin in feces of patients with
antimicrobial agent-associated diarrhea and miscellaneous conditions. J Clin Microbiol 15: 1049–
1053. PMID: 7107838
9.
Meyers S, Mayer L, Bottone E, Desmond E, Janowitz HD (1981) Occurrence of Clostridium difficile
toxin during the course of inflammatory bowel disease. Gastroenterology 80: 697–670. PMID:
7202942
10.
Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife WT, et al. (2008) Decreased diversity of the
fecal Microbiome in recurrent Clostridium difficile-associated diarrhea. J Infect Dis 197: 435–438. doi:
10.1086/525047 PMID: 18199029
11.
Aroniadis OC, Brandt LJ (2013) Fecal microbiota transplantation: past, present and future. Curr Opin
Gastroenterol 29: 79–84. doi: 10.1097/MOG.0b013e32835a4b3e PMID: 23041678
12.
Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, et al. (2006) An obesity-associated gut
microbiome with increased capacity for energy harvest. Nature 444: 1027–1031. PMID: 17183312
13.
Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, et al. (2009) A core gut microbiome
in obese and lean twins. Nature 457: 480–484. doi: 10.1038/nature07540 PMID: 19043404
14.
Qin J, Li Y, Cai Z, Li S, Zhu J, et al. (2012) A metagenome-wide association study of gut microbiota in
type 2 diabetes. Nature 490: 55–60. doi: 10.1038/nature11450 PMID: 23023125
15.
Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, et al. (2013) Microbiota modulate behavioral and
physiological abnormalities associated with neurodevelopmental disorders. Cell 155: 1451–1463. doi:
10.1016/j.cell.2013.11.024 PMID: 24315484
16.
Dapito DH, Mencin A, Gwak GY, Pradere JP, Jang MK, et al. (2012) Promotion of hepatocellular carci-
noma by the intestinal microbiota and TLR4. Cancer Cell 21: 504–516. doi: 10.1016/j.ccr.2012.02.007
PMID: 22516259
17.
Reddy BS, Watanabe K (1978) Effect of intestinal microflora on 2,2'-dimethyl-4-aminobiphenyl-induced
carcinogenesis in F344 rats. J Natl Cancer Inst 61: 1269–1271. PMID: 280712
18.
Bonneau R, Facciotti MT, Reiss DJ, Schmid AK, Pan M, et al. (2007) A predictive model for transcrip-
tional control of physiology in a free living cell. Cell 131: 1354–1365. PMID: 18160043
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
22 / 25
19.
Schmid AK, Reiss DJ, Kaur A, Pan M, King N, et al. (2007) The anatomy of microbial cell state transi-
tions in response to oxygen. Genome Res 17: 1399–1413. PMID: 17785531
20.
Thakar J, Pilione M, Kirimanjeswara G, Harvill ET, Albert R (2007) Modeling systems-level regulation of
host immune responses. PLoS Comput Biol 3: e109. PMID: 17559300
21.
Saez-Rodriguez J, Alexopoulos LG, Epperlein J, Samaga R, Lauffenburger DA, et al. (2009) Discrete
logic modelling as a means to link protein signalling networks with functional analysis of mammalian
signal transduction. Mol Syst Biol 5: 331. doi: 10.1038/msb.2009.87 PMID: 19953085
22.
Zhang R, Shah MV, Yang J, Nyland SB, Liu X, et al. (2008) Network model of survival signaling in large
granular lymphocyte leukemia. Proc Natl Acad Sci U S A 105: 16308–16313. doi: 10.1073/pnas.
0806447105 PMID: 18852469
23.
Saadatpour A, Wang RS, Liao A, Liu X, Loughran TP, et al. (2011) Dynamical and structural analysis of
a T cell survival network identifies novel candidate therapeutic targets for large granular lymphocyte
leukemia. PLoS Comput Biol 7: e1002267. doi: 10.1371/journal.pcbi.1002267 PMID: 22102804
24.
Stein RR, Bucci V, Toussaint NC, Buffie CG, Ratsch G, et al. (2013) Ecological modeling from time-
series inference: insight into dynamics and stability of intestinal microbiota. PLoS Comput Biol 9:
e1003388. doi: 10.1371/journal.pcbi.1003388 PMID: 24348232
25.
Marino S, Baxter NT, Huffnagle GB, Petrosino JF, Schloss PD (2014) Mathematical modeling of pri-
mary succession of murine intestinal microbiota. Proc Natl Acad Sci U S A 111: 439–444. doi: 10.1073/
pnas.1311322111 PMID: 24367073
26.
Davidich MI, Bornholdt S (2013) Boolean network model predicts knockout mutant phenotypes of fis-
sion yeast. PLoS One 8: e71786. doi: 10.1371/journal.pone.0071786 PMID: 24069138
27.
Bornholdt S (2005) Systems biology. Less is more in modeling large genetic networks. Science 310:
449–451. PMID: 16239464
28.
Steinway SN, Zanudo JG, Ding W, Rountree CB, Feith DJ, et al. (2014) Network modeling of TGFbeta
signaling in hepatocellular carcinoma epithelial-to-mesenchymal transition reveals joint sonic hedge-
hog and Wnt pathway activation. Cancer Res 74: 5963–5977. doi: 10.1158/0008-5472.CAN-14-0225
PMID: 25189528
29.
Naldi A, Monteiro PT, Mussel C, the Consortium for Logical M, Tools, et al. (2015) Cooperative develop-
ment of logical modelling standards and tools with CoLoMoTo. Bioinformatics.
30.
Campbell C, Yang S, Albert R, Shea K (2011) A network model for plant-pollinator community assem-
bly. Proc Natl Acad Sci U S A 108: 197–202. doi: 10.1073/pnas.1008204108 PMID: 21173234
31.
Oberhardt MA, Palsson BO, Papin JA (2009) Applications of genome-scale metabolic reconstructions.
Mol Syst Biol 5: 320. doi: 10.1038/msb.2009.77 PMID: 19888215
32.
Buffie CG, Jarchum I, Equinda M, Lipuma L, Gobourne A, et al. (2012) Profound alterations of intestinal
microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium diffi-
cile-induced colitis. Infect Immun 80: 62–73. doi: 10.1128/IAI.05496-11 PMID: 22006564
33.
Bartels RH, Beatty J.C., Barskey B.A. (1998) Hermite and Cubic Spline Interpolation. An Introduction to
Splines for Use in Computer Graphics and Geometric Modelling: Morgan Kaufmann. pp. 9–17.
34.
Saadatpour A, Albert R (2013) Boolean modeling of biological regulatory networks: a methodology tuto-
rial. Methods 62: 3–12. doi: 10.1016/j.ymeth.2012.10.012 PMID: 23142247
35.
Berestovsky N, Nakhleh L (2013) An Evaluation of Methods for Inferring Boolean Networks from Time-
Series Data. PLoS One 8: e66031. PMID: 23805196
36.
Lähdesmäki H. SI, Yli-Harja O (2003) On learning gene regulatory networks under the Boolean network
model. Machine Learning 52: 147–167.
37.
Mussel C, Hopfensitz M, Kestler HA (2010) BoolNet—an R package for generation, reconstruction and
analysis of Boolean networks. Bioinformatics 26: 1378–1380. doi: 10.1093/bioinformatics/btq124
PMID: 20378558
38.
Chaves M, Albert R, Sontag ED (2005) Robustness and fragility of Boolean models for genetic regula-
tory networks. J Theor Biol 235: 431–449. PMID: 15882705
39.
Albert I, Thakar J, Li S, Zhang R, Albert R (2008) Boolean network simulations for life scientists. Source
Code Biol Med 3: 16. doi: 10.1186/1751-0473-3-16 PMID: 19014577
40.
Aziz RK, Bartels D, Best AA, DeJongh M, Disz T, et al. (2008) The RAST Server: rapid annotations
using subsystems technology. BMC Genomics 9: 75. doi: 10.1186/1471-2164-9-75 PMID: 18261238
41.
Overbeek R, Olson R, Pusch GD, Olsen GJ, Davis JJ, et al. (2014) The SEED and the Rapid Annota-
tion of microbial genomes using Subsystems Technology (RAST). Nucleic Acids Res 42: D206–214.
doi: 10.1093/nar/gkt1226 PMID: 24293654
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
23 / 25
42.
Taffs R, Aston JE, Brileya K, Jay Z, Klatt CG, et al. (2009) In silico approaches to study mass and
energy flows in microbial consortia: a syntrophic case study. BMC Syst Biol 3: 114. doi: 10.1186/1752-
0509-3-114 PMID: 20003240
43.
Kanehisa M, Goto S, Sato Y, Kawashima M, Furumichi M, et al. (2014) Data, information, knowledge
and principle: back to metabolism in KEGG. Nucleic Acids Res 42: D199–205. doi: 10.1093/nar/
gkt1076 PMID: 24214961
44.
Kanehisa M, Goto S (2000) KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 28:
27–30. PMID: 10592173
45.
The MathWorks I (2012) MATLAB and Statistics Toolbox Release 2012b. Natick, Massachusetts,
United States.
46.
Levy R, Borenstein E (2013) Metabolic modeling of species interaction in the human microbiome eluci-
dates community-level assembly rules. Proc Natl Acad Sci U S A 110: 12804–12809. doi: 10.1073/
pnas.1300926110 PMID: 23858463
47.
Borenstein E, Kupiec M, Feldman MW, Ruppin E (2008) Large-scale reconstruction and phylogenetic
analysis of metabolic environments. Proc Natl Acad Sci U S A 105: 14482–14487. doi: 10.1073/pnas.
0806162105 PMID: 18787117
48.
Faust K, Raes J (2012) Microbial interactions: from networks to models. Nat Rev Microbiol 10: 538–
550. doi: 10.1038/nrmicro2832 PMID: 22796884
49.
Team RC (2013) R: A language and environment for statistical computing. R Foundation for Statistical
Computing. Vienna, Austria. doi: 10.3758/s13428-013-0330-5 PMID: 23519455
50.
Jensen PA, Dougherty BV, Moutinho TJ Jr., Papin JA (2015) Miniaturized plate readers for low-cost,
high-throughput phenotypic screening. J Lab Autom 20: 51–55. doi: 10.1177/2211068214555414
PMID: 25366331
51.
Deatherage Kaiser BL, Li J, Sanford JA, Kim YM, Kronewitter SR, et al. (2013) A Multi-Omic View of
Host-Pathogen-Commensal Interplay in-Mediated Intestinal Infection. PLoS One 8: e67155. PMID:
23840608
52.
Jump RL, Polinkovsky A, Hurless K, Sitzlar B, Eckart K, et al. (2014) Metabolomics analysis identifies
intestinal microbiota-derived biomarkers of colonization resistance in clindamycin-treated mice. PLoS
One 9: e101267. doi: 10.1371/journal.pone.0101267 PMID: 24988418
53.
Lawley TD, Clare S, Walker AW, Goulding D, Stabler RA, et al. (2009) Antibiotic treatment of clostrid-
ium difficile carrier mice triggers a supershedder state, spore-mediated transmission, and severe dis-
ease in immunocompromised hosts. Infect Immun 77: 3661–3669. doi: 10.1128/IAI.00558-09 PMID:
19564382
54.
Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, et al. (2015) Precision microbiome reconstitution
restores bile acid mediated resistance to Clostridium difficile. Nature 517: 205–208. doi: 10.1038/
nature13828 PMID: 25337874
55.
Ubeda C, Bucci V, Caballero S, Djukovic A, Toussaint NC, et al. (2013) Intestinal microbiota containing
Barnesiella species cures vancomycin-resistant Enterococcus faecium colonization. Infect Immun 81:
965–973. doi: 10.1128/IAI.01197-12 PMID: 23319552
56.
Lawley TD, Clare S, Walker AW, Stares MD, Connor TR, et al. (2012) Targeted restoration of the intesti-
nal microbiota with a simple, defined bacteriotherapy resolves relapsing Clostridium difficile disease in
mice. PLoS Pathog 8: e1002995. doi: 10.1371/journal.ppat.1002995 PMID: 23133377
57.
Reeves AE, Koenigsknecht MJ, Bergin IL, Young VB (2012) Suppression of Clostridium difficile in the
gastrointestinal tracts of germfree mice inoculated with a murine isolate from the family Lachnospira-
ceae. Infect Immun 80: 3786–3794. doi: 10.1128/IAI.00647-12 PMID: 22890996
58.
Klitgord N, Segre D (2010) Environments that induce synthetic microbial ecosystems. PLoS Comput
Biol 6: e1001002. doi: 10.1371/journal.pcbi.1001002 PMID: 21124952
59.
Sam Ma Z, Guan Q, Ye C, Zhang C, Foster JA, et al. (2015) Network analysis suggests a potentially
'evil' alliance of opportunistic pathogens inhibited by a cooperative network in human milk bacterial
communities. Sci Rep 5: 8275. doi: 10.1038/srep08275 PMID: 25651890
60.
Shankar V, Agans R, Holmes B, Raymer M, Paliy O (2013) Do gut microbial communities differ in pedi-
atric IBS and health? Gut Microbes 4: 347–352. doi: 10.4161/gmic.24827 PMID: 23674073
61.
Rigsbee L, Agans R, Shankar V, Kenche H, Khamis HJ, et al. (2012) Quantitative profiling of gut micro-
biota of children with diarrhea-predominant irritable bowel syndrome. Am J Gastroenterol 107: 1740–
1751. PMID: 22986438
62.
Trosvik P, de Muinck EJ, Stenseth NC (2014) Biotic interactions and temporal dynamics of the human
gastrointestinal microbiota. ISME J.
63.
Feist AM, Herrgard MJ, Thiele I, Reed JL, Palsson BO (2009) Reconstruction of biochemical networks
in microorganisms. Nat Rev Microbiol 7: 129–143. doi: 10.1038/nrmicro1949 PMID: 19116616
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
24 / 25
64.
Zomorrodi AR, Maranas CD (2012) OptCom: a multi-level optimization framework for the metabolic
modeling and analysis of microbial communities. PLoS Comput Biol 8: e1002363. doi: 10.1371/journal.
pcbi.1002363 PMID: 22319433
65.
Foster KR, Bell T (2012) Competition, not cooperation, dominates interactions among culturable micro-
bial species. Curr Biol 22: 1845–1850. doi: 10.1016/j.cub.2012.08.005 PMID: 22959348
66.
Pultz NJ, Stiefel U, Subramanyan S, Helfand MS, Donskey CJ (2005) Mechanisms by which anaerobic
microbiota inhibit the establishment in mice of intestinal colonization by vancomycin-resistant Entero-
coccus. J Infect Dis 191: 949–956. PMID: 15717271
67.
Buffie CG, Pamer EG (2013) Microbiota-mediated colonization resistance against intestinal pathogens.
Nat Rev Immunol 13: 790–801. doi: 10.1038/nri3535 PMID: 24096337
68.
Brandl K, Plitas G, Mihu CN, Ubeda C, Jia T, et al. (2008) Vancomycin-resistant enterococci exploit
antibiotic-induced innate immune deficits. Nature 455: 804–807. doi: 10.1038/nature07250 PMID:
18724361
69.
Kinnebrew MA, Ubeda C, Zenewicz LA, Smith N, Flavell RA, et al. (2010) Bacterial flagellin stimulates
Toll-like receptor 5-dependent defense against vancomycin-resistant Enterococcus infection. J Infect
Dis 201: 534–543. doi: 10.1086/650203 PMID: 20064069
Network Model of the Gut Microbiome
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004338
June 23, 2015
25 / 25
|
26102287
|
Lachnospiraceae = ( ( Lachnospiraceae ) AND NOT ( Clindamycin ) ) OR ( ( Lachnospiraceae_other ) AND NOT ( Clindamycin ) ) OR ( ( Other ) AND NOT ( Clindamycin ) )
Clindamycin = ( Clindamycin )
Akkermansia = ( Coprobacillus )
Enterobacteriaceae = ( Enterobacteriaceae )
Mollicutes = ( Mollicutes )
Barnesiella = ( ( Lachnospiraceae ) AND NOT ( Clindamycin ) ) OR ( ( Lachnospiraceae_other ) AND NOT ( Clindamycin ) ) OR ( ( Other ) AND NOT ( Clindamycin ) )
Lachnospiraceae_other = ( ( Lachnospiraceae ) AND NOT ( Clindamycin ) ) OR ( ( Other ) AND NOT ( Clindamycin ) ) OR ( ( Lachnospiraceae_other ) AND NOT ( Clindamycin ) )
Enterococcus = ( ( Mollicutes ) OR ( Blautia ) OR ( Enterobacteriaceae ) OR ( Clostridium_difficile ) ) OR NOT ( Mollicutes OR Clostridium_difficile OR Blautia OR Enterobacteriaceae OR Coprobacillus )
Clostridium_difficile = ( ( Clostridium_difficile ) AND NOT ( Barnesiella ) )
Blautia = ( Enterococcus ) OR ( Coprobacillus AND ( ( ( NOT Blautia ) ) ) ) OR ( Blautia AND ( ( ( NOT Coprobacillus ) ) ) )
Other = ( ( Lachnospiraceae ) AND NOT ( Clindamycin ) ) OR ( ( Lachnospiraceae_other ) AND NOT ( Clindamycin ) ) OR ( ( Other ) AND NOT ( Clindamycin ) )
|
1521-0103/354/3/448–458$25.00
http://dx.doi.org/10.1124/jpet.115.224766
THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS
J Pharmacol Exp Ther 354:448–458, September 2015
U.S. Government work not protected by U.S. copyright
Logic-Based and Cellular Pharmacodynamic Modeling of
Bortezomib Responses in U266 Human Myeloma Cellss
Vaishali L. Chudasama, Meric A. Ovacik, Darrell R. Abernethy, and Donald E. Mager
Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (V.L.C., M.A.O.,
D.E.M.); and Office of Clinical Pharmacology, Food and Drug Administration, Silver Springs, Maryland (D.R.A.)
Received March 28, 2015; accepted July 9, 2015
ABSTRACT
Systems models of biological networks show promise for in-
forming drug target selection/qualification, identifying lead com-
pounds and factors regulating disease progression, rationalizing
combinatorial regimens, and explaining sources of intersubject
variability and adverse drug reactions. However, most models of
biological systems are qualitative and are not easily coupled
with dynamical models of drug exposure-response relation-
ships. In this proof-of-concept study, logic-based modeling of
signal transduction pathways in U266 multiple myeloma (MM)
cells is used to guide the development of a simple dynamical
model linking bortezomib exposure to cellular outcomes.
Bortezomib is a commonly used first-line agent in MM
treatment; however, knowledge of the signal transduction
pathways regulating bortezomib-mediated cell cytotoxicity is
incomplete. A Boolean network model of 66 nodes was con-
structed that includes major survival and apoptotic pathways
and was updated using responses to several chemical probes.
Simulated responses to bortezomib were in good agreement
with experimental data, and a reduction algorithm was used to
identify key signaling proteins. Bortezomib-mediated apoptosis
was not associated with suppression of nuclear factor kB (NFkB)
protein inhibition in this cell line, which contradicts a major
hypothesis of bortezomib pharmacodynamics. A pharmacody-
namic model was developed that included three critical proteins
(phospho-NFkB, BclxL, and cleaved poly (ADP ribose) polymer-
ase). Model-fitted protein dynamics and cell proliferation profiles
agreed with experimental data, and the model-predicted IC50
(3.5 nM) is comparable to the experimental value (1.5 nM). The
cell-based pharmacodynamic model successfully links bortezomib
exposure to MM cellular proliferation via protein dynamics, and
this model may show utility in exploring bortezomib-based
combination regimens.
Introduction
The fields of systems biology and pharmacokinetic (PK)/
pharmacodynamic (PD) modeling have evolved largely in
parallel, and there is an emerging consensus that an effective
integration of these disciplines is needed to fully realize the
promise of each in bringing new therapeutic molecules and
combination regimens to the bedside (http://www.nigms.nih.
gov/Training/Documents/SystemsPharmaWPSorger2011.pdf).
Traditional PK/PD models of drug action use compartmen-
tal structures to integrate the time course of drug exposure,
pharmacological properties (capacity, sensitivity, and transduc-
tion of drug-target interactions), and (patho)physiological turn-
over processes (Mager et al., 2003). Such semimechanistic
models contain a minimal number of identifiable parameters
to describe temporal profiles of macroscale therapeutic and
adverse drug effects. When coupled with nonlinear mixed-
effects modeling of relatively large clinical trials, a covariate
analysis can be used to identify patient-specific characteristics
(e.g., genetic polymorphisms) that explain the interindividual
variability in model parameters (Pillai et al., 2005). Although
a major component of model-informed drug development and
therapeutics (Milligan et al., 2013), this approach can be limited
by specific study designs and is rarely sufficient for recapitu-
lating multiple, complex genotype-phenotype relationships.
Significant insights have been realized from the recognition
that both drugs and disease processes give rise to complex and
dynamic clinical phenotypes by altering natural intercon-
nected biochemical networks and support the emergence of
systems pharmacology models of drug action (Zhao and
Iyengar, 2012; Huang et al., 2013; Jusko, 2013). Multiscale
models that combine PK/PD principles and signaling net-
works can serve as a platform for integrating genomic/
proteomic factors that regulate drug effects and clinical out-
comes—so-called enhanced PD (ePD) models (Iyengar et al.,
2012). Two major challenges to the development of ePD
models include the lack of complete mathematical models of
signal transduction networks (e.g., concentrations and re-
action rate constants) and unknown quantitative relationships
This work was supported by the National Institutes of Health National
Institute of General Medical Sciences [Grant R01-GM57980] (to D.E.M.);
the University at Buffalo-Pfizer strategic alliance (D.E.M.); an unrestricted
training grant from Daiichi Sankyo Pharma Development (to D.E.M.); and an
American Foundation for Pharmaceutical Education predoctoral fellowship (to
V.L.C.).
dx.doi.org/10.1124/jpet.115.224766.
s This article has supplemental material available at jpet.aspetjournals.org.
ABBREVIATIONS: ePD, enhanced pharmacodynamics; FBS, fetal bovine serum; IkBi, inhibitor of kB inhibitor; IKKi, inhibitor of kB kinase inhibitor;
JAK, janus kinase; JNK, c-Jun N-terminal kinase; MM, multiple myeloma; NFkB, nuclear factor kB; PARP, poly (ADP ribose) polymerase; PD,
pharmacodynamics; pIkBa, phosphor-IkBa; PK, pharmacokinetics; pNFkB, phospho–nuclear factor kB; pStat3, phospho–signal transducer and
activator of transcription 3; RIP, receptor-interacting protein; STAT, signal transducer and activator of transcription.
448
between individual genomic/proteomic differences and model
parameters. Mechanistic network models are preferred over
empirical structures (Birtwistle et al., 2013); however, such
models may not be defined or calibrated to specific pharmaco-
logical and/or disease systems. Logic-based modeling tech-
niques provide a global perspective of system properties in
the absence of kinetic parameters through the integration of
qualitative a priori knowledge of network connections (Albert
and Wang, 2009). In this proof-of-concept study, a reduction
algorithm is applied to a mathematical network to guide the
development of a small signaling model for bortezomib, a potent
proteasome inhibitor, which may ultimately serve as an ePD
model for its use in multiple myeloma (MM).
Multiple myeloma is a B cell neoplasm associated with
several comorbidities, including hypercalcemia, renal insuffi-
ciency, anemia, and bone lesions (Caers et al., 2008; Blade
et al., 2010). The prognosis for advanced stages of MM is poor
despite multiple treatment options, with a median survival of
advanced-stage patients of less than 10 months (Richardson
et al., 2003, 2005). Bortezomib is commonly prescribed alone
or in combination with other antimyeloma agents (Oancea
et al., 2004), and the addition of bortezomib has significantly
improved overall survival of MM patients (Caers et al., 2008).
However, almost all patients relapse and become refractory to
all treatment options (Richardson et al., 2005; San Miguel
et al., 2008), and a better understanding of MM disease
progression and mechanisms of drug action is critical for
improving the treatment of MM.
Bortezomib modulates both survival and apoptotic cellular
pathways in MM cells (Fig. 1), and multiple mechanisms of
action are proposed for inducing cell death, including the
inhibition of proteasome and the phospho–nuclear factor kB
(pNFkB) pathway (Hideshima et al., 2001, 2002, 2003a,b).
However, recent reports suggest that bortezomib stimulates
receptor-interacting protein (RIP; a signaling protein up-
stream of the NFkB pathway), leading to activation of pNFkB
protein expression (Hideshima et al., 2009). Despite extensive
qualitative information on bortezomib-induced intracellular
signaling, mathematical models linking bortezomib exposure
to intracellular protein dynamics have not been established.
Systems-level modeling may facilitate the rational design of
single and combinatorial dosing regimens, approaches to
overcome drug resistance, and prevent suboptimal dosing
(Berger and Iyengar, 2011; Zhao and Iyengar, 2012). Mecha-
nistic models are emerging in which chemotherapy exposure is
connected to ultimate responses through target occupancy and
biomarker signal transduction (Yamazaki et al., 2011; Harrold
et al., 2012; Kay et al., 2012; Kirouac et al., 2013). The purpose
of this study is to integrate network systems analysis and PK/PD
modeling principles to study bortezomib effects on signal trans-
duction in MM cells. Although the current study is conducted in
U266 cells, the strategic framework may be extended to other cell
lines and applied to other therapeutic areas.
Materials and Methods
Tissue Culture Materials. The human myeloma cell line U266
(TIB-196) was purchased from American Type Culture Collection
(Manassas, VA). Inhibitor of kB kinase inhibitor (IKKi) (PS-1145) and
Janus kinase (JAK) inhibitor I (JAKi) were purchased from Santa
Cruz Biotechnology (Dallas, TX). IkBi (BAY11-7085) was purchased
from Sigma-Aldrich (St. Louis, MO). The clinically available formula-
tion of bortezomib was used for all experiments (Millennium Phar-
maceuticals, Cambridge, MA). The primary monoclonal antibodies to
phospho-p65, phosphor-IkBa (pIkBa), phospho–signal transducers
and activator of transcription 3 (pStat3), Bcl-xL, poly (ADP ribose)
polymerase (PARP), p65, Stat3, IkBa, and b-actin were purchased
from Cell Signaling Technology (Danvers, MA). a-Tubulin primary and
rabbit and mouse secondary antibodies were purchased from Santa Cruz
Biotechnology. WST-1 reagent assay kit was purchased from Roche
(Basel, Switzerland). Horseradish peroxidase conjugate was purchased
from Bio-Rad (Hercules, CA), and enhanced chemiluminescence sub-
strate was purchased from Thermo Scientific (Pittsburgh, PA).
Fig. 1. Signal transduction pathways in multiple my-
eloma disease pathology. Cyt-c, cytochrome c; DR4/5,
death receptor 4/5; FasL, TNF receptor superfamily
member 6 ligand; MAPK, mitogen-activated protein
kinase; NIK, NFkB-inducing kinase.
Mathematical Modeling of Cellular Responses to Bortezomib
449
RPMI 1640 and fetal bovine serum (FBS) were purchased from
American Type Culture Collection.
Cell Culture Experimental Design. All experiments were con-
ducted in U266 myeloma cells. Cells were cultured in RPMI 1640
medium supplemented with 15% FBS and 1% penicillin/streptomycin
antibiotics. Treatment protocols included 1) IKKi (10 mM), 2) JAKi
(10 mM), 3) IkBi (10 mM), 4) IKKi and JAKi (10 mM each), 5) IkBi and
JAKi (10 mM each), 6) bortezomib (20 and 2 nM), and 7) dimethylsulf-
oxide vehicle control. All treatments were continuous for 48 hours, with
the exception of IkBi experiments (10 hours). A transient exposure to
bortezomib (20 nM) was also conducted, in which cells were incubated for
either 1 or 2 hours followed by removal of drug from the culture media
and subsequent incubation with vehicle control for up to 48 hours.
WST-1 Cell Proliferation Assay. Approximately 10,000 cells per
well were incubated with a range of concentrations (0.001–100 mM) of
JAKi, IKKi, or a combination of IKKi and JAKi. Incubation experi-
ments with bortezomib alone ranged from 0.001 to 100 nM. Cell
viability was measured at 0, 24, 48, 72, and 96 hours according to
manufacturer instructions. Absorbance was measured at 450 and
690 nm after 2-hour incubation with WST-1 reagent with a Microplate
spectrophotometer (Molecular Devices, Sunnydale, CA).
Western Blot Analysis. Relative protein expression levels of
pStat3, pNFkB, pIkBa, cleaved PARP, and BclxL were measured
following treatment regimens as specified in Supplemental Table 1.
Cells (5 106/10 ml culture media) were incubated in 10-cm2 culture
dishes. Cells were collected at the end of the treatment duration, washed
with ice-cold phosphate-buffered saline, and lysed at 4°C in radio-
immunoprecipitation assay RIPA buffer (Cell Signaling Technology)
supplemented with protease and phosphatase inhibitor cocktail (Bio-
Rad) and phenylmethylsulfonyl fluoride (Bio-Rad). Protein lysates were
stored at 280°C until used. Equal amounts of proteins were separated
on SDS-PAGE gels and transferred to nitrocellulose membranes.
Immunoblotting was performed according to manufacturer instruc-
tions, and the relative intensity of bands was assessed by densito-
metric analysis of digitized images using ImageJ software (NIH,
Bethesda, MD). All experiments were conducted in at least duplicate.
Drug Stability. Bortezomib degradation in cell culture was
assessed at different time points (0, 12, 24, 48, and 72 hours).
Bortezomib concentrations were measured after incubation (20 nM) at
37°C in culture medium (RPMI 1640 supplemented with 15% FBS and
1% penicillin/streptomycin) using a liquid chromatography–tandem
mass spectrometry assay previously validated in our laboratory (un-
published). Bortezomib degradation kinetics were fit with a monoexpo-
nential (l) decay model, and the half-life was calculated as ln(2)/l.
Boolean Network Assembly and Simulations. The Boolean
network model (Supplemental Fig. 1) was drawn with SmartDraw
software (http://www.smartdraw.com) and was implemented in Odefy
(http://www.helmholtz-muenchen.de/cmb/odefy)
(Wittmann et
al.,
2009), a toolbox compatible with MATLAB (MathWorks, Natick,
MA). Odefy converts Boolean relationships into a continuous frame-
work using a Hill-type function. In the hypothetical case in which node
B stimulates A, the relationship can be described by
dA=dt 5 1=t½Bn=ðBn 1 knÞ 2 A :
Default parameter values for k (0.5), n (3), and t (1) were used for all
nodes. All model simulations were performed using the “HillCubeNorm”
option within Odefy (MATLAB model code is provided in the Data
Supplement). Each node in the model was initialized as ON (1) or OFF
(0) based on baseline activity in the cell. For example, if the state is
constitutively active in the cell, it was initialized as 1. Boolean transfer
functions and initial state values are summarized in Supplemental
Table 2. Examples of ordinary differential equations corresponding to
each node, which were generated using Odefy, are listed in Supplemental
Eqs. 1–5.
Three specific probes, JAKi, IkBi, and IKKi, were used to develop
and update the network model. The effect of the JAKi was added such
that it inhibits nodes JAK1 and JAK2. The IkBi inhibitor was added by
direct inhibition of pIkB node activation. Similarly, the IKKi effect was
added such that it inhibits node IKK. Model simulations were
performed to predict bortezomib outcomes, and drug effect was in-
corporated as direct inhibition of the proteasome node and stimulation
of the RIP node. For simulations of transient bortezomib exposure, the
bortezomib node was activated for only one or two iterations.
Network Reduction. The final full Boolean network model de-
veloped for U266 cells (Supplemental Fig. 1) was reduced using
a model reduction algorithm (Veliz-Cuba, 2011) to identify critical
proteins
involved
in
bortezomib-mediated
signal
transduction.
Figure 2 shows examples of the steps taken to reduce the network.
First, nodes with only one input and one output are eliminated and the
pathways are reconnected (Fig. 2A). For example, RAF was removed
and RAS and MEK1 were directly connected (Fig. 2B). In another
network reduction rule, nonfunctional edges are identified and re-
moved based on the Boolean relationships (algorithm S from
Veliz-Cuba, 2011). For example, Fig. 2C shows nodes A, B, and C,
with the following Boolean functions: f(A) 5 B OR (B AND NOT C), f(B)
5 (A AND C), and f(C) 5 A. Hence, node C is nonfunctional in f(A)
because of the “OR” relationship, and the B node is sufficient to elicit
the same effect without invoking C. The resulting diagram is shown
(block arrow, Fig. 2C) with the Boolean relationships: f(A) 5 B, f(B) 5
(A AND C), and f(C) 5 A. p21 is an example of nonfunctional edges
(Fig. 2D), and Boolean relationships for p21 [f(p21) 5 p53 AND (NOT
AKT OR NOT MDM OR NOT MYC OR NOT CKD4)] were refined
using step 2 to f(p21) 5 p53 AND NOT AKT to remove nonfunctional
edges. AKT is the sole connection to BAD; therefore, the p21
connection to AKT is retained (Fig. 2D).
Finally, nonfunctional nodes are identified and removed from the
network (algorithm R from Veliz-Cuba, 2011). Before applying this
rule, the network is rewired and the first rule is reapplied. Nodes that
do not have any impact on other nodes are considered nonfunctional
and are removed. In Fig. 2E, node C is regulating node B, but at the
same time node A is regulating node C. Therefore, the connection from
node C to B can be removed, and consequently node C can be removed.
As an example, the only node modulated by MYC is CYCE, as the MYC
connection to CYCD can be removed because of the AKT connection to
CYCD (Fig. 2F). pStat3, mitogen-activated protein kinase, and
extracellular signal-regulated kinase stimulate MYC independently,
and mitogen-activated protein kinase was removed following the first
network reduction rule (Fig. 2A). Similarly, extracellular signal-
regulated kinase was removed as it does not modulate any other node
besides MYC. pStat3 modulates several other nodes (e.g., BclxL and
XIAP) and stimulates MYC. MYC can be removed following the one
input–one output rule, and therefore, CYCE is directly modulated by
pStat3 (Fig. 2F), resulting in four final nodes. This entire process was
repeated until no further nodes could be removed from the Boolean
network. Boolean simulations of the final reduced network were
compared with simulations of the full network using Odefy (Wittmann
et al., 2009).
Dynamical Model Development. Several critical proteins iden-
tified using the model reduction algorithm were incorporated into
a reduced cellular PD model based on mechanisms of bortezomib action
and signal transduction pathways. The reduced dynamic model of
bortezomib is depicted in Fig. 3. In brief, bortezomib stimulates protein
expression of pNFkB and cleaved PARP (following delays) and inhibits
expression of BclxL. A transit compartment (M1B) was added to account
for the slight delay in stimulation of pNFkB protein expression (Mager
and Jusko, 2001). Bortezomib stimulates the synthesis rate of M1B
(ksyn_M1B) via a linear stimulation coefficient (Sm_M1B), and the rate of
change in expression of M1B is defined as follows:
dðM1BÞ
dt
5 ksyn M1B×
1 1 Sm_M1B×CB
2 kdeg_M1B×M1B; M1Bð0Þ 5 1
(1)
where CB is the concentration of bortezomib, and kdeg_M1B is a first-
order degradation rate constant of M1B. pNFkB protein expression
450
Chudasama et al.
increases but returns toward the baseline after 24 hours of continuous
bortezomib exposure. To capture this trend, a precursor-dependent
indirect response model was proposed (Sharma et al., 1998), with
stimulation of the first-order transfer rate constant from the precursor
pool (pre_pNFkB) to the pNFkB compartment (ktr_NFkB). The rate of
change of pre_pNFkB and pNFkB is described by the following
equations:
d
pre pNFkB
dt
5 ksyn_NFkB 2 ktr_NFkB × pre_pNFkB × M1B; pre_pNFkBð0Þ
5 ksyn NFkB
ktr NFkB
(2)
dðpNFkBÞ
dt
5 ktr_NFkB × pre_pNFkB × M1B
2 kdeg_NFkB × pNFkB; pNFkBð0Þ 5 1
(3)
where ksyn_NFkB is a zero-order production rate constant of pre_pNFkB,
and kdeg_NFkB is a first-order degradation rate constant of pNFkB.
Changes in cleaved PARP expression follow a significant delay, and
the protein expression profile was best characterized using a time-
dependent transduction model (Mager and Jusko, 2001), with negative
feedback from the last compartment to the synthesis rate of first cleaved
PARP compartment to characterize the downward phase of protein
expression. Bortezomib stimulates the synthesis rate (ksyn_PARP) of the
first cleaved PARP compartment (cPARP1) with a linear stimulation
coefficient (Sm_PARP):
dðcPARP1Þ
dt
5 ksyn_PARP
cPARP4
×
1 1 Sm_PARP_B × CB
2 ktr_PARP × cPARP1; cPARP1ð0Þ 5 1
(4).
The three subsequent transit compartments are described by the
following general equation:
dðcPARPnÞ
dt
5 ktr_PARP × ðcPARPn 2 1 2 cPARPnÞ; cPARPnð0Þ 5 1
(5)
with n representing the compartment number (n 5 2–4). pNFkB is
a prosurvival transcription factor that upregulates antiapoptotic
proteins (e.g., BclxL), whereas PARP is a proapoptotic factor that
results in apoptosis when activated (cleaved PARP) and stimulates
Fig. 2. Boolean network reduction steps. (A) Removal of nodes with one input and one output (see node Y). (B) Specific example of removing nodes with
one input and one output, in which nodes RAS and MEK1 are connected after removing RAF. (C) General example of identification and removal of
nonfunctional edges in a network, in which the edge from node C to node A is identified as a nonfunctional edge and removed. (D) Specific subnetwork
example of removing a nonfunctional edge for p21. (E) General example of identification and removal of nonfunctional nodes, in which node C is removed
after applying the rule in panel (A). (F) Specific subnetwork example for removing nonfunctional nodes (e.g., MYC). Block arrows indicate the final
network after applying the specific rule. “OR” relationship is represented by ‖, “AND” by &&, and “NOT” by ∼. ERK, extracellular signal-regulated
kinase; MEKK, mitogen-activated protein kinase.
Mathematical Modeling of Cellular Responses to Bortezomib
451
the degradation of BclxL. The rate of change of BclxL is defined as
follows:
dðBclxLÞ
dt
5 ksyn_Bcl × pNFkB 2 kdeg_Bcl × cPARPl
4 ×BclxL; BclxLð0Þ 5 1
(6)
where ksyn_Bcl is a zero-order synthesis rate constant of BclxL, kdeg_Bcl
is a first-order degradation rate constant of BclxL, and l is a power
coefficient for the cleaved PARP effect on BclxL. Cell proliferation is
dependent on the balance between apoptotic and antiapoptotic
signals. Therefore, cell proliferation (N) was modulated by BclxL
and cleaved PARP expression profiles, with the first-order natural cell
death rate constant (kd) inhibited by BclxL and stimulated by cleaved
PARP. N represents cell proliferation:
dðNÞ
dt 5 N ×
kg 2 kd×cPARP4
ð2-BclxLÞ
; Nð0Þ 5 1
(7)
with kg representing a first-order growth rate constant. Secondary
equations defining zero-order production rate constants are listed in
Supplemental Eqs. 6–9.
Data Analysis. The dynamical model (Fig. 3) was first fitted to
mean pNFkB and cleaved PARP data, followed by pooling of all
relative changes in these protein expression patterns (naïve pooled
data approach). Initially, pNFkB and cleaved PARP protein dynamics
were fit separately. BclxL data were subsequently included, and all of
the data including BclxL, pNFkB, and cleaved PARP protein dynamics
were fitted simultaneously. Next, cell proliferation was introduced to
the existing cellular dynamic model, and all of the data (three
biomarkers and cell proliferation) were fitted simultaneously. The in
vitro degradation half-life of bortezomib was included for all model
runs. Parameters were estimated using MATLAB (fminsearch func-
tion, maximum likelihood algorithm, and ode23s) and a model devel-
opment framework (Harrold and Abraham, 2014). All protein
dynamics were described using a proportional error variance model
(Yobs 5 Ypred×s), and cell proliferation was fitted using an additive plus
proportional error variance model (Yobs 5 Ypred×s 1 «), where Yobs is
the observation at time t, Ypred is the model-predicted value at time t,
and s and « are estimated variance model parameters.
Model Qualification. The final cell-based model and parameter
estimates were used to simulate protein dynamics and cell pro-
liferation after continuous exposure to a 10-fold-lower bortezomib
concentration (2 nM). Simulations were compared with the experi-
mentally measured cell proliferation and protein expression profiles.
Only parameters associated with natural cell proliferation (kg and kd)
were estimated. Cell proliferation was measured at 0, 24, 48, and
72 hours, and protein expression measurements were made at 0, 1, 4,
6, 8, 11, 24, 33, and 48 hours. The final model was also used to predict
cell proliferation at 48 hours for a range of bortezomib concentrations
(0.001–100 nM). The experimentally estimated IC50 value was
compared with the IC50 obtained from the simulated data.
Results
Network Development with IKK and JAK Inhibitors.
The final Boolean network model incorporates major survival
and apoptotic pathways in U266 cells (Supplemental Fig. 1),
and Supplemental Table 2 summarizes all node descriptions,
Boolean internodal relationships, and initial values. The
green connections in Supplemental Fig. 1 indicate how the
initial network was updated based on cellular responses to the
pathway probes. Nodes that are constitutively active under
baseline cell conditions were set as being active (1) or other-
wise inactive (0) in the network. An initial Boolean network
was constructed, and the performance of the initial model was
tested with two pathway-specific probes, IKKi and JAKi, to
inhibit the NFkB and JAK/STAT pathways. In the Boolean
network, the effect of each inhibitor was added such that IKKi
inhibits node IKK, and JAKi inhibits nodes JAK1 and JAK2.
Simulations of the initial Boolean network show that nodes
representing pIkBa, pNFkB, pStat3, and BclxL expression
decrease gradually upon inhibition of node IKK (Supplemen-
tal Fig. 2A, black dash-dotted lines). Inhibition of pStat3
expression appears to be due to interleukin-6 inhibition (major
stimulus of JAK/STAT3 pathway) via pNFkB expression.
However, in vitro Western blot experiments show that
expressions of pNFkB, pStat3, and BclxL remained un-
changed after 48-hour exposure to the IKKi, whereas pIkBa
expression decreased (Supplemental Fig. 2B, black symbols
and dotted lines). In contrast, simulations of the same proteins
using the initial model following JAKi exposure (Supplemen-
tal Fig. 2A, green dashed lines) were comparable to observed
experimental profiles over 48 hours (Supplemental Fig. 2B,
green symbols and dashed lines). To account for the observed
trend in pNFkB expression, the initial network was modified
such that pStat3 also stimulates pNFkB (Supplemental Fig. 1).
Although not yet confirmed in U266 or other MM cell lines,
studies suggest cross-talk between NFkB and STAT3 signaling
in tumors (Squarize et al., 2006; Lee et al., 2009; Saez-Rodriguez
et al., 2011). To further test this hypothesis, model simulations
were compared for the combination of IKKi and JAKi treatment
with measured temporal expression profiles of pNFkB. Simu-
lations of pNFkB following combination treatment with JAKi
and IKKi using the initial Boolean network showed a steady
decrease over time (data not shown). However, pNFkB expres-
sion measured by Western blot analysis only transiently de-
creased with a return to baseline values (Supplemental Fig. 2B,
blue symbols and dash-dot lines). A detailed evaluation of
pNFkB is described in the next section. Overall, cellular protein
dynamic simulations using the final model (Supplemental Fig.
2C) reasonably agree with the experimental results (Supple-
mental Fig. 2B).
Fig. 3. Signal transduction model of bortezomib effects in multiple
myeloma. Bortezomib compartment represents bortezomib concentration
in vitro with a degradation half-life of 144 hours. M1B is a signaling
compartment for stimulating the conversion of the precursor compart-
ment (pre_NFkB) to pNFkB, which represents relative pNFkB protein
expression. cPARPn represents transit compartment n for cleaved PARP.
The Bcl-xL compartment represents relative BclxL protein expression,
and N is cell proliferation in vitro. Yellow highlighted boxes are
compartments for which experimental data are measured. Open and
closed rectangles represent stimulation and inhibition processes.
452
Chudasama et al.
NFkB Dynamics. As the expression of pNFkB was not
suppressed by the IKKi, either as a single agent or in
combination with the JAKi, cellular responses to another
NFkB pathway inhibitor (IkBi) were evaluated alone or in
combination with the JAKi. Using the initial Boolean net-
work, inhibition of node pIkBa by IkBi resulted in simulations
showing inhibition of pNFkB expression (data not shown).
However, continuous IkBi exposure to U266 cells for up to
10 hours (alone and in combination with JAKi) maintained
pIkBa expression below baseline values (Supplemental Fig.
3A), whereas pNFkB expression transiently decreased fol-
lowed by a steady increase above the baseline (Supplemental
Fig. 3A). Based on the lack of pNFkB suppression with either
the IKKi or IkBi, alone or in combination with JAKi,
additional factors were assumed to govern pNFkB expression
in U266 cells outside of IKK, pIkBa, and pStat3 signaling.
This phenomenon was emulated by incorporating a dummy
node “X” to constantly stimulate pNFkB in the final network
(Supplemental Fig. 1). The addition of this factor stabilized
the Boolean network and reconciled simulations (Supplemen-
tal Fig. 2, C, D, and F) with all experimental data (Supple-
mental Fig. 2, B, E, and G). These results highlight the need to
inhibit NFkB activity directly rather than through upstream
pathways. Both final model simulations and experimental
data confirm inhibition of pIkBa expression following contin-
uous 48-hour exposure to the IKKi (10 mM), as well as the lack
of suppression of pNFkB and BclxL expression levels (Sup-
plemental Fig. 2, B and C, black symbols and dotted lines).
Analogously, continuous exposure to the JAKi (10 mM) sup-
pressed expression of pStat3, whereas pNFkB and BclxL
expression levels remained unchanged (Supplemental Fig. 2,
B and C, green symbols and dashed lines). In addition, final
model simulations of U266 cellular proliferation and apoptosis
agreed well with experimental cellular responses (Supple-
mental Fig. 2, D–G). A range of IKKi and JAKi concentrations
(0.001–100 mM) was used to evaluate cell viability, and
neither induction of apoptosis nor decreased cell viability
was observed after 72-hour continuous exposure alone or in
combination (Supplemental Fig. 2, E and G).
Bortezomib Pharmacodynamics. Once the final Bool-
ean network model was updated using protein dynamics and
cellular responses after exposures to probe inhibitors, the
model was used to evaluate bortezomib pharmacodynamics.
Bortezomib effect was introduced into the network such that it
directly inhibits the “proteasome” node and stimulates the
“RIP” node (Supplemental Fig. 1). Stimulation of RIP acti-
vates a downstream cascade, leading to activation of pIkBa
followed by pNFkB. In the Boolean network, a state initialized
as 1 (active) cannot be further stimulated; therefore, pIkBa
and pNFkB signal intensities remain unchanged (Supplemen-
tal Fig. 4A, blue dash-dot line). However, our immunoblot
analysis revealed transient increases in pNFkB and pIkBa
expression levels, followed by a gradual return to baseline
values (Supplemental Fig. 4B, blue symbols and lines), which
is in agreement with published profiles (Hideshima et al.,
2009). Comparing steady-state values for pNFkB and pIkBa
before and after treatment suggests no change in expression
levels, which is in agreement with Boolean network simula-
tions (Supplemental Fig. 4A). Furthermore, inhibition of
proteasome results in accumulated cellular stress that acti-
vates the c-Jun N-terminal kinase (JNK) pathway as well as
the apoptotic pathway. The mitochondrial apoptotic pathway
activates caspase-3, leading to activation of p53 followed by
p21, resulting in the inhibition of the Bcl-2 family proteins
(e.g., BclxL and Bcl-2) (Supplemental Fig. 4B, blue symbols
and lines). The activated JNK pathway inversely regulates
pStat3, leading to downregulation of pStat3 expression (Sup-
plemental Fig. 4B, blue symbols and lines). Simulations using
the final Boolean network model correctly predicted the
inhibition of pStat3 and BclxL in U266 cells after bortezomib
exposure (Supplemental Fig. 4A, blue symbols and lines). In
addition, final model simulations of U266 cellular outcomes to
bortezomib exposure were in good agreement with in vitro
measurements of cell viability and apoptosis measured via
cleaved PARP expression (Fig. 4).
To further test the fidelity of the Boolean network model to
predict bortezomib pharmacodynamics, we compared Boolean
network simulations with experimental results of cellular
responses upon transient bortezomib exposures. Simulations
of transient drug exposure were achieved by maintaining the
Bortezomib node as active (1 5 ON) for a limited number of
iterations, and the dynamics and steady states of protein and
cellular response nodes were monitored. Transient experi-
ments were conducted in which U266 cells were briefly
exposed (i.e., for 1 and 2 hours) to bortezomib. Interestingly,
some nodes (e.g., proteasome) returned to baseline values once
bortezomib was removed from the system, whereas other
nodes (e.g., cleaved PARP or Cl. PARP) achieved a new steady
state even after drug was removed. Furthermore, steady-state
outcomes for several nodes varied depending on the duration
of simulated bortezomib exposure. For example, a relatively
short duration of proteasome suppression produces a transient
induction of apoptosis, whereas longer durations of bortezomib
exposure resulted in steady states of apoptosis induction and
inhibited cell growth that are similar to simulated outcomes
Fig. 4. Comparison of model-simulated and observed U266 cellular
outcomes with continuous bortezomib exposure. (A) Cell proliferation
over 96 hours after bortezomib (20 nM) exposure in U266 cells. (B) Time
course of apoptosis as measured by cleaved PARP protein expression
under constant exposure to bortezomib (20 nM) in U266 cells. (C) Final
Boolean model (Supplemental Fig. 1) simulations of cell proliferation. (D)
Apoptosis under control conditions (red, solid) and after constant exposure
to bortezomib (blue, dash-dot line). Symbols are the mean observed data,
and error bars represent the standard deviation.
Mathematical Modeling of Cellular Responses to Bortezomib
453
following continuous drug exposure. A shorter duration of
bortezomib exposure was associated with a delay in apoptosis
induction as compared with continuous drug exposure (Supple-
mental Fig. 5A), and similar trends were observed experimen-
tally (Supplemental Fig. 5B). Western blot analysis of BclxL
and cleaved PARP expression shows a reduced magnitude in
changes from baseline values between transient and continu-
ous bortezomib exposure. The magnitude of cleaved PARP
induction was reduced from 22- to 6-fold after 24 hours of
bortezomib treatment. Model simulations of selected proteins
were comparable with experimental results (Supplemental Fig.
5, A and B). However, stimulation of cleaved PARP is associated
with apoptosis induction, downregulation of total NFkB ex-
pression, and subsequent suppression of pNFkB expression. In
contrast, incubation of U266 cells with bortezomib (20 nM) for
2 hours failed to inhibit pNFkB expression (Supplemental Fig.
5C, bottom), despite activation of cleaved PARP for over
48 hours (Supplemental Fig. 5B, apoptosis, black triangles). A
specific threshold of cleaved PARP activation might be required
to inhibit NFkB expression, which is not incorporated into the
current network model, and further studies are needed to test
this hypothesis.
Network Reduction. A Boolean network reduction ap-
proach (Veliz-Cuba, 2011) identified eight critical nodes
(Supplemental Fig. 6A), five of which are survival pathway
nodes (AKT, pNFkB, BclxL, proteasome, and pRB) and three
are apoptotic pathway nodes (caspase-3, JNK, and p21).
Boolean simulations of the reduced network are identical to
the steady-state values of the full network, which further
supports the reduction algorithm (Supplemental Fig. 6B).
Although caspase-3 was identified as a critical protein through
the Boolean network reduction, cleaved PARP expression was
measured as a marker for apoptosis due to caspase-3 assay
limitations.
Bortezomib Cellular Pharmacodynamic Model. Three
critical proteins (i.e., pNFkB, BclxL, and cleaved PARP) were
selected from the reduced Boolean network model (Supplemen-
tal Fig. 6A) for measurement and inclusion in the cellular
pharmacodynamic model. The final cellular model (Fig. 3)
integrates the time courses of bortezomib exposure, protein
dynamics, and cell proliferation. Bortezomib elicits its effects on
pNFkB via stimulation of upstream proteins in the pNFkB
pathway and on cleaved PARP via stress accumulation due to
proteasome inhibition. Upon continuous exposure of U266 cells
to bortezomib, pNFkB protein expression is stimulated after
a slight delay with a return toward the baseline (Fig. 5A). The
slight delay in the stimulation of pNFkB was well characterized
by adding a simple transit compartment (M1B; Eqs. 1 and 2).
pNFkB expression was well described by a precursor model, in
which the delayed signal (M1B) stimulates the first-order
transfer from the precursor to the pNFkB compartment (Eqs.
2 and 3). Parameters associated with M1B and pNFkB (kdeg_M1B
and Sm_M1B) were not estimated with good precision; therefore,
M1B-associated parameters were fixed to the estimates during
an initial model run. This had no impact on model performance
(i.e., model fits were not compromised and parameter values did
not change substantially), but precision of the estimated
parameters was significantly improved (Table 1). A relatively
long delay was observed before cleaved PARP expression
increased (Fig. 5B), and a transit-compartment model was
selected to describe this delay. Four transit compartments were
found to be adequate, and the total transit time was well
estimated at 26.8 hours (Table 1, ktr_parp). BclxL expression
starts decreasing after 12 hours (Fig. 5C), and this delay was
also well characterized by the proposed model. The initial phase
of BclxL was maintained at steady state by pNFkB, and the
eventual decrease in BclxL expression was successfully de-
scribed using cleaved PARP stimulation of the BclxL degrada-
tion rate constant. An exponential growth model well captured
the natural cell proliferation under control conditions (Fig. 5D,
triangles). The final signal transduction model reasonably
captured the delay in cell death, and parameters were esti-
mated with good precision (Table 1). Overall, the pharmacody-
namic model well characterized the time courses of protein
expression and cell proliferation profiles after exposure to
bortezomib at 20 nM.
Sensitivity to Dose and Model Qualification. The final
pharmacodynamic model (Fig. 3) was developed using several
cellular biomarkers after continuous exposure to a single,
relatively high concentration of bortezomib (20 nM). Simu-
lations of protein expression profiles and cell proliferation
following exposure to different bortezomib concentrations
(0.1–20 nM) were performed to evaluate the role of bortezomib
dose in cellular outcomes (Supplemental Fig. 7). As bortezomib
concentration is increased, the magnitude of pNFkB expression
is also increased, and the time to peak response is decreased
(Supplemental Fig. 7A). The extent of induced cleaved PARP is
also increased (Supplemental Fig. 7B), along with greater
suppression of BclxL expression (Supplemental Fig. 7C) as
bortezomib concentration increases. For cell proliferation, cell
growth is suppressed up to 2 nM (Supplemental Fig. 7D, red
line), but greater concentrations result in cell death (Supple-
mental Fig. 7D). As an external predictive check, protein ex-
pression profiles and cell proliferation after low-dose bortezomib
exposure (2 nM) were measured and compared with model
simulations, which were obtained using the final model (Fig. 3)
and parameter estimates (Table 1) from the high-dose
bortezomib (20 nM) experiments. The low concentration of
bortezomib (2 nM) was chosen, as it is close to its IC50 value
(1.5 nM) from the in vitro exposure-response relationship at
48 hours. Thus, the selected concentration is relatively low
but should still elicit a pharmacological response. Model-
predicted pNFkB, cleaved PARP, and BclxL profiles and cell
proliferation dynamics are in good agreement with the
experimental data (Supplemental Fig. 8).
The final model was also used to predict the IC50 for
bortezomib after 48 hours of continuous exposure. Although
the model was developed based on temporal profiles after
a single concentration of bortezomib, the model-predicted IC50
was comparable to that obtained from the in vitro concentration-
effect experiments (3.5 versus 1.5 nM; Fig. 6).
Discussion
Network-based approaches are increasingly used in drug
discovery and development (Csermely et al., 2013; Harrold
et al., 2013). Pharmacological networks are now commonly
used for target identification (Sahin et al., 2009), evaluating
signaling networks between normal and diseased conditions
(Saez-Rodriguez et al., 2011), and understanding interactions
among pathways (Thakar et al., 2007; Ge and Qian, 2009; Mai
and Liu, 2009). For example, a Boolean network was used to
identify novel targets to overcome trastuzumab resistance in
breast cancer cell lines (Sahin et al., 2009), in which c-MYC
454
Chudasama et al.
was identified as a potential therapeutic target. In another
example, Boolean network modeling was used to analyze
immune responses due to the presence of virulence factors in
lower respiratory tract infection (Thakar et al., 2007). Three
distinct phases of Bordetellae infection were identified, which
was not possible using traditional experimental methods
(i.e., biochemical and molecular biology techniques). A similar
approach is used in the present analysis, in which a Boolean
network model is applied for evaluating bortezomib signal
transduction pathways in U266 cells.
Our initial Boolean network model was developed based on
the literature; however, available studies focus on either
NFkB (Bharti et al., 2003b) or JAK/STAT3 (Bharti et al.,
2003a; Park et al., 2011) pathways independently, and joint
effects of these pathways have not been previously evaluated
in U266 cells (e.g., apoptosis and cell proliferation). In
addition, cellular protein dynamics and outcomes following
exposure to specific probe compounds were also unavailable.
Therefore, in vitro experiments and computational modeling
were combined with pathway-specific inhibitors to evaluate
Fig. 5. Time course of cellular protein dynamics and
cell proliferation after continuous bortezomib exposure
(20 nM). Relative protein expression profiles are shown
for pNFkB (A), cleaved PARP (B), and BclxL (C). Protein
expression was measured using Western blot in whole-
cell lysate. (D) Cell proliferation measured using WST-1
assay kit. Measurements following bortezomib treat-
ment and vehicle control are represented by circles and
triangles. Solid lines are model-fitted profiles.
TABLE 1
Final Bortezomib parameter estimates in U266 cells
Parameter
Unit
Definition
Value
%CV
Protein dynamic parameters
ktr_NFkB
h21
pNFkB transit rate constant
0.000429a
—
kdeg_NFkB
h21
pNFkB degradation rate constant
0.54
52.0
kp_NFkB
h21
pNFkB precursor transfer rate constant
0.034
5.76
Sm_NFkB
nM21
pNFkB stimulatory coefficient
24.8a
—
ktr_parp
h21
cPARP transit rate constant
0.149
4.11
Sm_parp
nM21
cPARP stimulatory coefficient
6.72
11.4
Ybcl
—
BclxL power coefficient
0.949
14.6
kdeg_bcl
h21
BclxL degradation rate constant
7.72E-03
58.4
«NFkB
—
pNFkB proportional error coefficient
0.097
11.4
«bcl
—
BclxL proportional error coefficient
0.163
11.1
«parp
—
cPARP proportional error coefficient
0.229
11.7
In vitro cell dynamic parameters
kg
h21
Cell growth rate constant
0.021
3.74
kd
h21
Cell death rate constant
2.56E-03
9.81
C0cell
Initial cell density
1a
—
scell
—
Cell additive error coefficient
0.031
32.9
«cell
—
Cell proportional error coefficient
0.157
21.7
%CV, percent coefficient of variation.
aFixed value.
Mathematical Modeling of Cellular Responses to Bortezomib
455
the roles of both pathways individually and simultaneously.
The Boolean network model confirmed that stress accumula-
tion due to proteasome inhibition is a major pathway of
myeloma cell death despite the activation of pNFkB protein
expression, which contradicts a major proposed hypothesis of
bortezomib effects.
Whereas logic-based models provide qualitative insight into
network connectivity and drug-induced signal transduction,
a quantitative dynamical model that includes important
cellular biomarkers allows for the integration of critical
factors responsible for cell death upon chemotherapy exposure
(Yamazaki et al., 2011; Kay et al., 2012; Zhang et al., 2013). It
is not yet practical to quantify all components in cellular
signaling pathways; therefore, it is necessary to identify
critical proteins for dynamical model development. A Boolean
network reduction algorithm (Veliz-Cuba, 2011) identified
critical factors regulating bortezomib cell death that guided
development of a reduced PD model. Among the biomarkers
(Supplemental Fig. 6), two antiapoptotic proteins and one
proapoptotic protein (pNFkB, BclxL, and cleaved PARP) were
integrated into a reduced pharmacodynamic model (Fig. 3).
pNFkB was integrated in the model as bortezomib increases
its expression, contradictory to proposed mechanisms. BclxL
was selected as a major antiapoptotic protein that inhibits
apoptosis, and cleaved PARP is a primary proapoptotic
marker. The factor p21 was not incorporated in the current
model as Boolean simulations suggest that bortezomib effects
on apoptosis precede cell growth arrest.
The final cellular PD model (Fig. 3) was successfully
qualified using the external data set following a low concen-
tration of bortezomib (2 nM). Despite the fact that the cellular
model was developed based on a single bortezomib concentra-
tion (20 nM) and linear coefficients, the final model reasonably
predicted responses to the lower bortezomib exposure (Sup-
plemental Fig. 8) and the dose-response curve for bortezomib
at 48 hours (Fig. 6). Although there is a slight misfit for BclxL
and cleaved PARP profiles (Supplemental Fig. 8, B and C),
overall simulated profiles reasonably agree with experimental
data. Most commonly, exposure-response relationships of
protein expression data are sigmoidal in nature (Bharti
et al., 2003a,b; Park et al., 2008, 2011). For example, the
inhibition of pNFkB activity by curcumin is 10% at 1 mM and
40% at 10 mM (Bharti et al., 2003b). Similarly, cleaved PARP
activation is about 1% at 10 mM curcumin but 100% at 50 mM
curcumin (Park et al., 2008). It is therefore interesting that,
although our model was developed using a single drug
concentration and simple linear coefficients (e.g., Sm_M1B
and Sm_PARP in Eqs. 1 and 4), it reasonably predicts profiles
at a lower bortezomib concentration (Supplemental Fig. 8) and
a sigmoidal concentration-effect relationship (Fig. 6). Further-
more, the predicted exposure-response relationship for BclxL
can be overlaid with observed cell proliferation at 48 hours
(Supplemental Fig. 9). This suggests that BclxL could serve as
a potential biomarker to predict efficacy in MM and warrants
further study. However, a potential limitation is that the
proteins measured in this study reflect relative fold change
and not absolute values of protein concentrations. Grounding
on relative changes in protein expression maintains a degree
of modularity, but more advanced proteomic methods are
needed to quantitatively measure low-abundance signaling
proteins. Cell growth and death parameters were also com-
pared with available values from the literature. Natural cell
growth and death were modeled using first-order growth rate
constants (kg and kd). The estimated net growth rate constant
(kg – kd) in the final model (0.00490 h21) is comparable to net
growth rate constants from U266 xenografts at 0.00487 h21
(Siveen et al., 2014) and 0.00450 h21 (Rhee et al., 2012) that
were obtained by fitting an exponential model to digitized
control curves (data not shown).
Preliminary
experiments
were
conducted
to
measure
pNFkB in the nucleus and cytoplasm of U266 cells after
bortezomib exposure. Both cellular fractions showed a similar
trend of increased relative pNFkB expression (data not
shown); therefore, total cellular pNFkB was measured by
Western blot analysis of cell lysates, revealing transient
increased expression (Fig. 5A). Despite the stimulation of
pNFkB protein expression, apoptosis is induced and the model
is able to well capture this phenomenon. The NFkB pathway is
one of the major cell survival pathways and is deregulated in
many types of cancer (Karin, 2009). Bortezomib stimulates the
upstream cascade of the NFkB pathway (Hideshima et al.,
2009) leading to activation of pNFkB protein expression.
Apoptosis is still induced through cellular stress resulting
from the inhibition of proteasome (Fig. 4D) (Hideshima et al.,
2001). The cellular model was able to well describe both
mechanisms. Aberrant expression of pNFkB is responsible for
the lack of efficacy and resistance in some cases of MM
(Hideshima et al., 2002; Bharti et al., 2004). Greater expres-
sion of pNFkB results in greater expression of antiapoptotic
proteins (e.g., BclxL) (Karin and Lin, 2002), with cross-talk
between survival and apoptotic pathways determining cellu-
lar fate. Hence, greater drug concentrations are required to
activate the apoptotic pathway. Initially, BclxL inhibition of
cleaved PARP activation and cleaved PARP stimulation on
removal of BclxL was incorporated. However, since BclxL
exerts a negative feedback on cell death, as incorporated in the
model, the BclxL effect on cleaved PARP was removed to
simplify the model and reduce model redundancy.
In summary, systems pharmacology is an emerging field
that seeks to couple systems biology and PD modeling, which
could promote the discovery, development, and effective use of
drugs based upon first principles. Computational tools based
on graph theory (Liu et al., 2013), discrete dynamic relation-
ships (Albert and Wang, 2009), and others can be used to
identify critical system components within networks at mul-
tiple organizational levels, thus providing guidance for
Fig.
6. Comparison of model-predicted (Pred) and observed (Obs)
bortezomib concentration-effect profiles at 48 hours. Circles represent
mean observed data (error bars are S.D.) at 48 hours. Lines represent
either a fitted curve to observed data using a Hill-type function (red) or the
model-predicted data at 48 hours (blue).
456
Chudasama et al.
multiscale model construction and evaluation. We have used
an integrated approach to investigate bortezomib signal
transduction and exposure-response relationships in MM
cells. A cell-based model of bortezomib in U266 human
myeloma cells was developed that incorporates one of the
major survival pathway proteins (pNFkB), antiapoptotic pro-
tein BclxL, and an apoptotic marker (cleaved PARP) to link
bortezomib exposure to cell proliferation. The final model-
predicted in vitro IC50 for bortezomib reasonably agrees with
the experimental IC50 at 48 hours. Although the Boolean and
dynamical models are relatively simple and specific to U266
cells and bortezomib, the overall strategic approach can be
easily extended with slight modification to other myeloma cell
lines (e.g., MM.1S and RPMI 8226) or bortezomib-based
combination regimens (e.g., histone deacetylase inhibitors).
This model may serve as a basis for studying bortezomib
combinations with antimyeloma agents and to optimize
xenograft combination studies, which is a focus of current
research and will be reported separately. In addition, the
model structure can be easily extended to describe responses
in other cancer types that share similar pathways.
Acknowledgments
The authors thank Dr. John M. Harrold (University at Buffalo,
SUNY) for developing MATLAB code for this project.
Authorship Contributions
Participated in research design: Design and execution of experi-
ments: Chudasama.
Conducted experiments: Chudasama.
Performed data analysis: Chudasama, Ovacik, Mager.
Wrote or contributed to the writing of the manuscript: Chudasama,
Ovacik, Abernethy, Mager.
References
Albert R and Wang RS (2009) Discrete dynamic modeling of cellular signaling net-
works. Methods Enzymol 467:281–306.
Berger SI and Iyengar R (2011) Role of systems pharmacology in understanding drug
adverse events. Wiley Interdiscip Rev Syst Biol Med 3:129–135.
Bharti AC, Donato N, and Aggarwal BB (2003a) Curcumin (diferuloylmethane)
inhibits constitutive and IL-6-inducible STAT3 phosphorylation in human multiple
myeloma cells. J Immunol 171:3863–3871.
Bharti AC, Donato N, Singh S, and Aggarwal BB (2003b) Curcumin (diferuloylmethane)
down-regulates the constitutive activation of nuclear factor-kappa B and IkappaBalpha
kinase in human multiple myeloma cells, leading to suppression of proliferation and
induction of apoptosis. Blood 101:1053–1062.
Bharti AC, Shishodia S, Reuben JM, Weber D, Alexanian R, Raj-Vadhan S, Estrov Z,
Talpaz M, and Aggarwal BB (2004) Nuclear factor-kappaB and STAT3 are con-
stitutively active in CD1381 cells derived from multiple myeloma patients, and
suppression of these transcription factors leads to apoptosis. Blood 103:3175–3184.
Birtwistle MR, Mager DE, and Gallo JM (2013) Mechanistic vs. Empirical network
models of drug action. CPT Pharmacometrics Syst Pharmacol 2:e72.
Bladé J, Cibeira MT, Fernández de Larrea C, and Rosiñol L (2010) Multiple myeloma.
Ann Oncol 21 (Suppl 7):vii313–vii319.
Caers J, Vande broek I, De Raeve H, Michaux L, Trullemans F, Schots R, Van Camp
B, and Vanderkerken K (2008) Multiple myeloma–an update on diagnosis and
treatment. Eur J Haematol 81:329–343.
Csermely P, Korcsmáros T, Kiss HJ, London G, and Nussinov R (2013) Structure and
dynamics of molecular networks: a novel paradigm of drug discovery: a compre-
hensive review. Pharmacol Ther 138:333–408.
Ge H and Qian M (2009) Boolean network approach to negative feedback loops of the
p53 pathways: synchronized dynamics and stochastic limit cycles. J Comput Biol
16:119–132.
Harrold JM and Abraham AK (2014) Ubiquity: a framework for physiological/
mechanism-based pharmacokinetic/pharmacodynamic model development and
deployment. J Pharmacokinet Pharmacodyn 41:141–151.
Harrold JM, Ramanathan M, and Mager DE (2013) Network-based approaches in
drug discovery and early development. Clin Pharmacol Ther 94:651–658.
Harrold JM, Straubinger RM, and Mager DE (2012) Combinatorial chemotherapeutic
efficacy in non-Hodgkin lymphoma can be predicted by a signaling model of CD20
pharmacodynamics. Cancer Res 72:1632–1641.
Hideshima T, Chauhan D, Hayashi T, Akiyama M, Mitsiades N, Mitsiades C, Podar
K, Munshi NC, Richardson PG, and Anderson KC (2003a) Proteasome inhibitor
PS-341 abrogates IL-6 triggered signaling cascades via caspase-dependent down-
regulation of gp130 in multiple myeloma. Oncogene 22:8386–8393.
Hideshima T, Chauhan D, Richardson P, Mitsiades C, Mitsiades N, Hayashi T,
Munshi N, Dang L, Castro A, Palombella V, et al. (2002) NF-kappa B as a thera-
peutic target in multiple myeloma. J Biol Chem 277:16639–16647.
Hideshima T, Ikeda H, Chauhan D, Okawa Y, Raje N, Podar K, Mitsiades C, Munshi
NC, Richardson PG, Carrasco RD, et al. (2009) Bortezomib induces canonical nu-
clear factor-kappaB activation in multiple myeloma cells. Blood 114:1046–1052.
Hideshima T, Mitsiades C, Akiyama M, Hayashi T, Chauhan D, Richardson P,
Schlossman R, Podar K, Munshi NC, Mitsiades N, et al. (2003b) Molecular
mechanisms mediating antimyeloma activity of proteasome inhibitor PS-341.
Blood 101:1530–1534.
Hideshima T, Richardson P, Chauhan D, Palombella VJ, Elliott PJ, Adams J,
and Anderson KC (2001) The proteasome inhibitor PS-341 inhibits growth, induces
apoptosis, and overcomes drug resistance in human multiple myeloma cells.
Cancer Res 61:3071–3076.
Huang SM, Abernethy DR, Wang Y, Zhao P, and Zineh I (2013) The utility of mod-
eling and simulation in drug development and regulatory review. J Pharm Sci 102:
2912–2923.
Iyengar R, Zhao S, Chung SW, Mager DE, and Gallo JM (2012) Merging systems
biology with pharmacodynamics. Sci Transl Med 4:126ps7.
Jusko WJ (2013) Moving from basic toward systems pharmacodynamic models.
J Pharm Sci 102:2930–2940.
Karin M (2009) NF-kappaB as a critical link between inflammation and cancer. Cold
Spring Harb Perspect Biol 1:a000141.
Karin M and Lin A (2002) NF-kappaB at the crossroads of life and death. Nat
Immunol 3:221–227.
Kay BP, Hsu CP, Lu JF, Sun YN, Bai S, Xin Y, and D’Argenio DZ (2012)
Intracellular-signaling tumor-regression modeling of the pro-apoptotic receptor
agonists dulanermin and conatumumab. J Pharmacokinet Pharmacodyn 39:
577–590.
Kirouac DC, Du JY, Lahdenranta J, Overland R, Yarar D, Paragas V, Pace E,
McDonagh CF, Nielsen UB, and Onsum MD (2013) Computational modeling of
ERBB2-amplified breast cancer identifies combined ErbB2/3 blockade as superior
to the combination of MEK and AKT inhibitors. Sci Signal 6:ra68.
Lee H, Herrmann A, Deng JH, Kujawski M, Niu G, Li Z, Forman S, Jove R,
Pardoll DM, and Yu H (2009) Persistently activated Stat3 maintains consti-
tutive NF-kappaB activity in tumors. Cancer Cell 15:283–293.
Liu YY, Slotine JJ, and Barabási AL (2013) Observability of complex systems. Proc
Natl Acad Sci USA 110:2460–2465.
Mager DE and Jusko WJ (2001) Pharmacodynamic modeling of time-dependent
transduction systems. Clin Pharmacol Ther 70:210–216.
Mager DE, Wyska E, and Jusko WJ (2003) Diversity of mechanism-based pharma-
codynamic models. Drug Metab Dispos 31:510–518.
Mai Z and Liu H (2009) Boolean network-based analysis of the apoptosis network:
irreversible apoptosis and stable surviving. J Theor Biol 259:760–769.
Milligan PA, Brown MJ, Marchant B, Martin SW, van der Graaf PH, Benson N,
Nucci G, Nichols DJ, Boyd RA, Mandema JW, et al. (2013) Model-based drug
development: a rational approach to efficiently accelerate drug development. Clin
Pharmacol Ther 93:502–514.
Oancea M, Mani A, Hussein MA, and Almasan A (2004) Apoptosis of multiple my-
eloma. Int J Hematol 80:224–231.
Park J, Ayyappan V, Bae EK, Lee C, Kim BS, Kim BK, Lee YY, Ahn KS, and Yoon SS
(2008) Curcumin in combination with bortezomib synergistically induced apoptosis
in human multiple myeloma U266 cells. Mol Oncol 2:317–326.
Park S, Lee HJ, Jeong SJ, Song HS, Kim M, Lee HJ, Lee EO, Kim DH, Ahn KS,
and Kim SH (2011) Inhibition of JAK1/STAT3 signaling mediates compound
K-induced apoptosis in human multiple myeloma U266 cells. Food Chem Toxicol
49:1367–1372.
Pillai GC, Mentré F, and Steimer JL (2005) Non-linear mixed effects modeling - from
methodology and software development to driving implementation in drug devel-
opment science. J Pharmacokinet Pharmacodyn 32:161–183.
Rhee YH, Jeong SJ, Lee HJ, Lee HJ, Koh W, Jung JH, Kim SH, and Sung-Hoon K
(2012) Inhibition of STAT3 signaling and induction of SHP1 mediate anti-
angiogenic and antitumor activities of ergosterol peroxide in U266 multiple mye-
loma cells. BMC Cancer 12:28.
Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D, Rajkumar
SV, Srkalovic G, Alsina M, Alexanian R, et al. (2003) A phase 2 study of bortezomib
in relapsed, refractory myeloma. N Engl J Med 348:2609–2617.
Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T,
Harousseau JL, Ben-Yehuda D, Lonial S, Goldschmidt H, et al.; Assessment of
Proteasome Inhibition for Extending Remissions (APEX) Investigators (2005)
Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl
J Med 352:2487–2498.
Saez-Rodriguez J, Alexopoulos LG, Zhang M, Morris MK, Lauffenburger DA,
and Sorger PK (2011) Comparing signaling networks between normal and trans-
formed hepatocytes using discrete logical models. Cancer Res 71:5400–5411.
Sahin O, Fröhlich H, Löbke C, Korf U, Burmester S, Majety M, Mattern J,
Schupp I, Chaouiya C, Thieffry D, et al. (2009) Modeling ERBB receptor-
regulated G1/S transition to find novel targets for de novo trastuzumab re-
sistance. BMC Syst Biol 3:1.
San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M,
Spicka I, Petrucci MT, Palumbo A, Samoilova OS, et al.; VISTA Trial Investigators
(2008) Bortezomib plus melphalan and prednisone for initial treatment of multiple
myeloma. N Engl J Med 359:906–917.
Sharma A, Ebling WF, and Jusko WJ (1998) Precursor-dependent indirect pharma-
codynamic response model for tolerance and rebound phenomena. J Pharm Sci 87:
1577–1584.
Siveen KS, Mustafa N, Li F, Kannaiyan R, Ahn KS, Kumar AP, Chng WJ, and Sethi
G (2014) Thymoquinone overcomes chemoresistance and enhances the anticancer
effects of bortezomib through abrogation of NF-kB regulated gene products in
multiple myeloma xenograft mouse model. Oncotarget 5:634–648.
Mathematical Modeling of Cellular Responses to Bortezomib
457
Squarize CH, Castilho RM, Sriuranpong V, Pinto DS, Jr, and Gutkind JS (2006)
Molecular cross-talk between the NFkappaB and STAT3 signaling pathways in
head and neck squamous cell carcinoma. Neoplasia 8:733–746.
Thakar J, Pilione M, Kirimanjeswara G, Harvill ET, and Albert R (2007)
Modeling systems-level regulation of host immune responses. PLOS Comput
Biol 3:e109.
Veliz-Cuba A (2011) Reduction of Boolean network models. J Theor Biol 289:
167–172.
Wittmann DM, Krumsiek J, Saez-Rodriguez J, Lauffenburger DA, Klamt S,
and Theis FJ (2009) Transforming Boolean models to continuous models: meth-
odology and application to T-cell receptor signaling. BMC Syst Biol 3:98.
Yamazaki S, Nguyen L, Vekich S, Shen Z, Yin MJ, Mehta PP, Kung PP, and Vicini
P (2011) Pharmacokinetic-pharmacodynamic modeling of biomarker response
and tumor growth inhibition to an orally available heat shock protein 90
inhibitor in a human tumor xenograft mouse model. J Pharmacol Exp Ther 338:
964–973.
Zhang T, Li Y, Zou P, Yu JY, McEachern D, Wang S, and Sun D (2013) Physiologi-
cally based pharmacokinetic and pharmacodynamic modeling of an antagonist
(SM-406/AT-406) of multiple inhibitor of apoptosis proteins (IAPs) in a mouse xe-
nograft model of human breast cancer. Biopharm Drug Dispos 34:348–359.
Zhao S and Iyengar R (2012) Systems pharmacology: network analysis to identify
multiscale mechanisms of drug action. Annu Rev Pharmacol Toxicol 52:505–521.
Address
correspondence
to:
Dr.
Donald
E.
Mager,
Department
of
Pharmaceutical Sciences, University at Buffalo, SUNY, 431 Kapoor Hall,
Buffalo, NY 14214. E-mail: dmager@buffalo.edu
458
Chudasama et al.
|
26163548
|
FLIP = ( pNFKB )
Apo = ( Cl_PARP )
cJun = ( JNK )
Prot = NOT ( ( Bort ) )
MEK1 = ( RAF )
XIAP = ( ( ( pSTAT3 ) AND NOT ( p53 ) ) AND NOT ( Smac ) ) OR ( ( ( pNFKB ) AND NOT ( p53 ) ) AND NOT ( Smac ) )
CYCE = ( MYC )
DNAdam = ( STRESS ) OR ( Cas3 )
BCL2 = ( ( ( pSTAT3 ) AND NOT ( p53 ) ) AND NOT ( BAD ) ) OR ( ( ( pNFKB ) AND NOT ( p53 ) ) AND NOT ( BAD ) )
p27 = ( ( ( ( p53 ) AND NOT ( AKT ) ) AND NOT ( MYC ) ) AND NOT ( CDK4 ) )
CYCD = ( AKT ) OR ( MYC ) OR ( ERK )
IKK = ( AKT ) OR ( RIP AND ( ( ( NIK ) ) ) )
pSTAT3 = ( ( JAK2 AND ( ( ( JAK1 AND STAT3 ) AND ( ( ( NOT IKK ) ) ) ) ) ) AND NOT ( JNK ) ) OR ( JAK1 AND ( ( ( STAT3 AND JAK2 ) ) ) )
CDK4 = ( ( ( CYCD ) AND NOT ( p27 ) ) AND NOT ( p21 ) )
Bclxl = ( ( ( ( pSTAT3 ) AND NOT ( p53 ) ) AND NOT ( BAD ) ) AND NOT ( BAX ) ) OR ( ( ( ( pNFKB ) AND NOT ( p53 ) ) AND NOT ( BAD ) ) AND NOT ( BAX ) )
JAK1 = ( gp130 AND ( ( ( IL6 ) ) ) )
BAD = NOT ( ( AKT ) )
CDK2 = ( ( CYCE ) AND NOT ( p21 ) )
MYC = ( pSTAT3 ) OR ( MEKK ) OR ( ERK )
ERK = ( MAPK )
PIP3 = ( ( PIP3 ) AND NOT ( PTEN ) )
MITO = ( BAX )
MEKK2 = ( RAC )
JAK2 = ( ( IL6 AND ( ( ( gp130 ) ) ) ) AND NOT ( SHP1 ) )
PI3K = ( IL6 AND ( ( ( gp130 ) ) ) )
JNK = ( ( MKK4 ) AND NOT ( Prot ) )
Cas3 = ( ( Cas8 ) AND NOT ( XIAP ) ) OR ( ( Cas9 ) AND NOT ( XIAP ) )
Cytc = ( MITO )
pRB = ( CDK2 AND ( ( ( CDK6 AND CDK4 ) ) ) ) OR ( CDK4 AND ( ( ( CDK6 ) ) ) )
Fas = ( FasL ) OR ( p53 )
gp130 = NOT ( ( Cas3 ) )
MKK4 = ( MEKK2 )
STRESS = ( ( DNAdam ) ) OR NOT ( DNAdam OR Prot )
IL6 = ( pNFKB )
RAC = ( STRESS )
MDM = ( ( AKT ) AND NOT ( ATM ) ) OR ( ( p53 ) AND NOT ( ATM ) )
pNFKB = ( pSTAT3 ) OR ( X ) OR ( pIKB ) OR ( Prot AND ( ( ( pIKB ) ) ) )
TRAF3 = NOT ( ( CIAP ) )
AKT = ( PIP3 )
CIAP = ( TNFAR )
Smac = ( MITO )
MAPK = ( MEK1 )
pIKB = ( IKK )
Cas8 = ( ( Fas AND ( ( ( FasL ) ) ) ) AND NOT ( FLIP ) )
GROWTH = ( pRB )
STAT3 = NOT ( ( Cas3 ) )
NIK = NOT ( ( TRAF3 ) )
p21 = ( ( ( ( ( p53 ) AND NOT ( MYC ) ) AND NOT ( AKT ) ) AND NOT ( CDK4 ) ) AND NOT ( MDM ) )
RAS = ( SHP1 ) OR ( IL6 AND ( ( ( gp130 ) ) ) )
ATM = ( DNAdam ) OR ( Cas3 )
p53 = ( ( JNK ) AND NOT ( MDM AND ( ( ( Prot ) ) ) ) ) OR ( ( DNAPK ) AND NOT ( MDM AND ( ( ( Prot ) ) ) ) )
BID = ( ( ( STRESS ) AND NOT ( BCL2 ) ) AND NOT ( Bclxl ) ) OR ( ( ( Cas8 ) AND NOT ( BCL2 ) ) AND NOT ( Bclxl ) ) OR ( ( ( Fas ) AND NOT ( BCL2 ) ) AND NOT ( Bclxl ) )
RIP = ( Bort ) OR ( TNFAR AND ( ( ( TNFA ) ) ) )
BAX = ( ( ( BID ) AND NOT ( Bclxl ) ) AND NOT ( BCL2 ) ) OR ( ( ( p53 ) AND NOT ( Bclxl ) ) AND NOT ( BCL2 ) )
FasL = ( cJun ) OR ( Fas )
RAF = ( RAS )
Cl_PARP = ( Cas3 )
CDK6 = ( CYCD )
MEKK = ( MEK1 )
PTEN = ( p53 )
DNAPK = ( ATM )
Cas9 = ( ( ( Cytc ) AND NOT ( XIAP ) ) AND NOT ( AKT ) )
|
RESEARCH ARTICLE
Predicting Variabilities in Cardiac Gene
Expression with a Boolean Network
Incorporating Uncertainty
Melanie Grieb2,5☯, Andre Burkovski2,3,5☯, J. Eric Sträng2☯, Johann M. Kraus2,
Alexander Groß1, Günther Palm3, Michael Kühl4*, Hans A. Kestler1,2,3*
1 Leibniz Institute for Age Research, Fritz-Lipmann Institute, Jena, Germany, 2 Core Unit Medical Systems
Biology, Ulm University, Ulm, Germany, 3 Neural Information Processing, Ulm University, Ulm, Germany,
4 Institute for Biochemistry and Molecular Biology, Ulm University, Ulm, Germany, 5 International Graduate
School of Molecular Medicine, Ulm University, Ulm, Germany
☯These authors contributed equally to this work.
* michael.kuehl@uni-ulm.de (MK); hkestler@fli-leibniz.de (HAK)
Abstract
Gene interactions in cells can be represented by gene regulatory networks. A Boolean net-
work models gene interactions according to rules where gene expression is represented by
binary values (on / off or {1, 0}). In reality, however, the gene’s state can have multiple val-
ues due to biological properties. Furthermore, the noisy nature of the experimental design
results in uncertainty about a state of the gene. Here we present a new Boolean network
paradigm to allow intermediate values on the interval [0, 1]. As in the Boolean network, fixed
points or attractors of such a model correspond to biological phenotypes or states. We use
our new extension of the Boolean network paradigm to model gene expression in first and
second heart field lineages which are cardiac progenitor cell populations involved in early
vertebrate heart development. By this we are able to predict additional biological pheno-
types that the Boolean model alone is not able to identify without utilizing additional biologi-
cal knowledge. The additional phenotypes predicted by the model were confirmed by
published biological experiments. Furthermore, the new method predicts gene expression
propensities for modelled but yet to be analyzed genes.
Introduction
Specialization of cells during development and differentiation is driven by transcription or
growth factors. These are interconnected in gene regulatory networks. The temporary regu-
lated interaction of these factors are finally resulting in terminally differentiated, specialized
cells which are characterized by the expression of a certain set of genes. Thus, development and
function of a certain cell type is largely reflected by the expression of selected genes in a cell.
Gene regulatory networks describe the interactions between those genes in the cell [1–3].
During embryonic development, these gene regulatory networks evolve over time towards a
stable state, finally reflecting the terminally differentiated cell [1], i.e., biological phenotypes.
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
1 / 15
OPEN ACCESS
Citation: Grieb M, Burkovski A, Sträng JE, Kraus JM,
Groß A, Palm G, et al. (2015) Predicting Variabilities
in Cardiac Gene Expression with a Boolean Network
Incorporating Uncertainty. PLoS ONE 10(7):
e0131832. doi:10.1371/journal.pone.0131832
Editor: Lars Kaderali, Technische Universität
Dresden, Medical Faculty, GERMANY
Received: March 5, 2015
Accepted: June 6, 2015
Published: July 24, 2015
Copyright: © 2015 Grieb et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Funding: This work was funded in part by the
German federal ministry of education and research
(BMBF) within the framework GERONTOSYS II
(Forschungskern SyStaR, Project ID 0315894A to
MK and HAK), European Community’s Seventh
Framework Programme (FP7/2007-2013) under grant
agreement no. 602783 (to HAK), and the International
Graduate School in Molecular Medicine at Ulm
University (GSC270). The funders had no role in
study design, data collection and analysis, decision to
publish, or preparation of the manuscript.
A gene regulatory network can be visualized as a static map that describes the interaction of
these genes and reflects the activation or inactivation of genes by other factors in the network.
Such a gene regulatory network can be implemented as a Boolean network if one assumes that
a gene can be either active or inactive in a cell and thus can be represented by a Boolean value
(on / off or {1,0}). Interaction between genes can then be mathematically modeled by Boolean
functions. A set of such logical rules or functions, more exactly one Boolean function per con-
sidered gene defines a Boolean network (BN) [4, 5]. Given some initial expression pattern, a
BN computes the evolution of gene expression in discrete time steps. Of particular importance
are states which are invariant or lead to periodic sequences of expression patterns, so called
attractors. For finite sized BNs any initial state will converge to one of these attractors in finite
time [6] In a Boolean network representing a gene regulatory network, these attractors are the
equivalent to the stable state of gene expression reflecting the differentiated biological pheno-
type of the cell.
BNs are useful as a first approach when it comes to model complex networks with many
genes and their interactions [7]. Often the BN is modeled from known regulatory interactions
that are manually derived from qualitative wet-lab experiments [8] or computationally deter-
mined with BN reconstruction methods [9, 10]. Additionally, simulated Boolean states of
genes from the simulation allow an intuitive interpretation of the results. Recently, BN models
have been used to capture the essence of gene regulation in several biological processes such as
the mammalian cell cycle [11], the guard cell abscisic acid signaling [12], or the oxidative stress
response pathway [13].
Modelling of gene regulatory networks and their simulation, however, is hampered by dif-
ferent drawbacks. In practice, for example, absolute data for gene expression activities are mea-
sured indirectly, e.g., by quantifying the relative amounts of the corresponding transcripts.
These measurements are inherently noisy. Furthermore, some notion of activity/inactivity has
to be inferred in order to infer the state of the gene. To this effect binarization schemes are used
in order to differentiate between active and inactive genes in time series data [14]. Here, one
also has to consider that effective thresholds are gene dependent [8]. Finally, one has to take
into account that gene expression can vary between different cells of an apparently homoge-
neous population of cells as previously shown for the common cardiac progenitor cell popula-
tion that gives rise to the heart [15].
Here, we implement a novel extension of the Boolean network paradigm and illustrate the
procedure on a Heart Field Development model. We also illustrate the utility of the method on
the Mammalian Cell Cycle [11] (see Section G in S1 Supplementary Information File). Our pri-
mary focus is the evolutionary conserved core cardiac regulatory network that drives early car-
diac development and that predicts the dynamic behaviour of gene expression during early
development [16]. In this process a common cardiac progenitor cell population splits into two
populations of cells called first (FHF) and second heart field (SHF), that are characterized by
the selective expression of typical transcription factors such as Isl1, Tbx1, Tbx5, and Nkx2.5.
Cells of the FHF develop to the primary heart tube and later to the left ventricle and the atria,
whereas cells of the SHF mainly develop into the right ventricle and the outflow tract [17].
Activation of these genes during cardiac development is regulated among others through
growth factors of the Wnt family [18]. The BN model of this gene regulatory network correctly
predicts the general pattern of gene expression in general, work in Xenopus [19] and in mouse
ES cells [15] albeit suggests that during cardiac development on a single cell level a much more
complex variability in gene expression exists.
In an attempt to include the variability of single cell gene expression in an otherwise homog-
enous cell population, our goal was to describe gene expression levels with multiple values,
extending the binary values of a BN. Previously, transformations of BNs have also been
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
2 / 15
Competing Interests: The authors have declared
that no competing interests exist.
considered by others [20, 21]. An overview over these approaches can be found in [22, 23].
These transform the Boolean rules into a system of ordinary differential equations which
describe the dynamics of gene concentrations. As opposed to the analysis of behaviour and
evolution of concentration levels we use an approach to model continuous intervals of gene
expression values in order to potentially find new fixed points using only the original BN. In
our model, we call continuous values attributed to genes “propensities” of gene expression and
do not need additional parameters that are otherwise required for modelling concentrations.
The corresponding interactions are derived from the Boolean functions of the BN. The opera-
tions AND, OR, and NOT are replaced by their arithmetic counterparts based on fuzzy logic
product sum rules [24]. This extends the BN into a discrete-time, non-linear, dynamical model
that is represented by a system of difference equations, namely, a BN extension (BNE). Natu-
rally, fixed points of this new model also represent possible expression patterns or phenotypes
of cells. Interestingly, this novel method is able to predict variabilities in gene expression during
cardiac development and cell cycle (see Figs M and N in S1 Supplementary Information File).
For cardiac development the extension predicts additional phenotypes that are in agreement
with published results and novel gene expression pattern for yet to be analyzed genes.
Methods
Since we intertwine biological and mathematical terms we shortly present an overview of the
important terminology that we use in the following:
• Boolean fixed point: A fixed point of the BN. It takes values Bn 2 {0,1}n with n being the
number of variables in the BN.
• BNE fixed point: A fixed point of the BNE. It takes values In 2 [0, 1]n.
• biological phenotype: A phenotype that describes a binarized gene expression of measure-
ments. It takes values in Bn, i.e., in form of binary gene expression.
• hypothetical phenotype: A pattern of gene expression. Hypothetical phenotypes are used in
order to map the extension fixed points to their nearest neighbouring pattern. In our particu-
lar case we consider binary gene expression patterns with values in Bn (a set of 2n binary
patterns).
The extension and analysis of the BN is conducted in several steps drafted in Fig 1 and are
described in detail below. Given a BN, using the canonical Disjunctive Normal Form (DNF),
each term is then transformed into a sum of products. The extended model is then simulated
to find BNE fixed points of the model. These fixed points can be interpreted by a mapping the
fixed points to the nearest hypothetical phenotypes. The meaning of the nearest hypothetical
phenotype must then be further interpreted in biological context.
Boolean Networks (BN)
BN were pioneered by Kauffman in 1969 [25, 26] to model gene interactions in a cell. BNs are
based on the assumption that a cell regulates its function in a time-dependent manner by
switching genes on (active) or off (inactive). The regulatory mechanism is described by logical
rules where the state of a gene is defined by logic rules based on the previous gene states.
Here, we denote by B = {0, 1} the set of Boolean values in binary representation. Formally, a
BN is defined by n Boolean variables x = (x1, . . ., xn) 2 Bn and a vector of n Boolean transition
functions F = (f1, . . ., fn), which describe the interaction between variables.
A transition function fi is a map fi:Bn ! B. In that way that the discrete time evolution gen-
erated by a BN is defined by sequential application of the vector valued function F = (f1, . . ., fn)
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
3 / 15
on an initial state x at time 0. A state transition is thus formally described by the map
xðt þ 1Þ ¼ FðxðtÞÞ; t 2 N0;
ð1Þ
given the state x(t) at time t. Bn is finite and discrete and there are 2n unique states. Each is a
Fig 1. Phenotype analysis using the Boolean Network Extension (BNE). The application of the BNE to a
given Boolean network (BN) can be divided into three basic steps: Model extension (top), identification of
stable structures (middle) and mapping to phenotypes (bottom). In the model extension step (top) the rules of
the BN are transformed to the rules of the BNE by converting the rules of the BN to canonical disjunctive
normal form (DNF) and then to product-sum fuzzy logic (DNF product-sum extension, details see section
Extension of Boolean networks). In the “identification of stable structures”-step (middle) the extended BNE is
simulated for a large number of random inputs. The resulting approximated attractors can either be fixed
points approximated by point clouds, fixed points depending on one or multiple parameters or different
dependencies. Finally, new phenotypes are identified step (bottom) by mapping the fixed points to their
nearest hypothetical phenotype.
doi:10.1371/journal.pone.0131832.g001
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
4 / 15
priori allowed as BN initial value. It follows that evolution of an initial state will converge to a
cyclic sequence of states in finite time. Such sequences are called attractors. Attractors are usu-
ally categorized in
• fixed points (attractors of period 1) and
• periodic sequences (attractors of period T > 1).
In the following synchronous BN will be considered. In synchronous BN, each Boolean func-
tion is executed once at each time step.
In a Boolean network any variable may interact with any subset of variables in the network.
Additionally, variables with constant values may be considered in order to parametrize external
inputs, e.g., influences of exogenous factors. Discrete time delays may also be modeled by intro-
ducing a chain of “dummy” variables which consecutively take the value of the chain’s first var-
iable value. Such a chain of n additional “dummy” variables result in a time delay of n steps of
the considered variable.
Boolean Network Extension (BNE)
By I we denote the interval [0, 1]. The new model extends the state space from Bn to In. We do
so by adapting the logical rules of the original BN. In general one has to find a mapping from
the Boolean operators AND (^), OR (_), and NOT (¬) to continuous operators (functions) on
In. For the sake of consistency, these operators should render the same results as their corre-
sponding Boolean analogue when restricting the values to {0, 1}.
A common approach is to treat the Boolean transition functions as fuzzy logic functions
[24, 27]. In the fuzzy logic literature, there are mainly two approaches. The first approach is the
min–max fuzzy logic. The second approach, product–sum fuzzy logic, replaces the Boolean
algebraic operators on Bn (_, ^,¬) with their arithmetic counterparts on In, namely, (+, ×) and
negation by (1 −x). Constant values of the BN are translated to constant functions of the BNE
from I. Contrarily to the min–max fuzzy logic, the product–sum fuzzy logic are differentiable
functions. Additionally, product–sum allows a smooth evolution and interaction between the
variables which correspond the idea that interactions in cells depend on the concentration lev-
els of products. In contrary, in min–max variables that are not minimal or maximal do not
influence the resulting value. In the following, we will only consider the product–sum fuzzy
logic.
Formally, the extension of a Boolean function
fi : Bn ! B
ð2Þ
a7!bi
ð3Þ
is the function
^f i : In ! I
ð4Þ
^a 7! ^bi;
ð5Þ
i.e., we consider the extensions of the Boolean domains and functions given by the operationb.
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
5 / 15
The product–sum fuzzy logic results in the following properties
c
:xi ¼ 1 ^xi;
ð6Þ
b
xi ^ xj ¼ ^xi ^xj;
ð7Þ
b
xi _ xj ¼ ^xi þ ^xj ^xi ^xj:
ð8Þ
Eq 8 can be derived from Eqs 6 and 7 using DeMorgan’s law. The Boolean formulae are trans-
formed into a unique representation using the canonical disjunctive normal form (DNF)
which can be directly derived from the truth table. Then, given a Boolean formula in DNF, we
directly extend it into the continuous version by applying the product-sum rules. The resulting
formula have values in I since DNF encodes a truth table for every possible term with n vari-
ables [28]. It is easily verified that the BN and its extension coincide for variable values in Bn
when applying the DNF/product–sum extension (DNFPS).
Example: sequence of transformations for the Boolean function f(a, b, c) = (a_b)^c,
Booleanfunction
!
DNF
!
DNFPS
ða _ bÞ ^ c
!
ð:a ^ b ^ cÞ _ ða ^ :b ^ cÞ _ ða ^ b ^ cÞ
!
ð^a þ ^b ^a ^bÞ ^c
Extension fixed points
As in the case of BN, its extension will have fixed points. Fixed points of the model are meant
to represent biological states since both are stable and time-invariant. Firstly, we defined the
extension procedure in such a way that a BN and its extension are consistent when restricting
to values in Bn. Hence, the BN fixed points are fixed points of its extension as well. Secondly,
an extension may have non–Boolean fixed points.
One crucial point of the extension is the fact that the variables of the system, i.e., variables
which are considered as constant input, may now take values in I. If these parameters are
uncertain, unknown, or the behavior of the system is to be examined, one would have to inves-
tigate the variations of the fixed points as the input variables are changed. Compared to the BN
case, two new aspects need to be taken into account. It is a priori not possible to predict the
number of fixed points of a BNE nor is it possible to conduct an exhaustive search. Addition-
ally, the iteration of a BNE can only be (practically) carried out numerically. Thus the values of
the fixed points given by iteration are not exact. The same would be true if the fixed points
were found by numerically solving ^f ^x
ð Þ ¼ ^x. Any attempt to systematically find the set of
extension fixed points of an extension would hence result in a numerical approximation of
fixed points gained by sweeping the initial values of the respective search through In. Amongst
those found, several may correspond to a single actual fixed point. A proper identification of
the true fixed point is therefore necessary. They can, e.g., be grouped into fixed point proto-
types by using k-means. The fixed points of the BNE would typically describe surfaces over the
investigated parameter space (see Fig 2).
Numerical considerations
BN were simulated with the help of the BoolNet package [29]. Fixed points were investigated in
two manners
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
6 / 15
• iteration with initial data taken from uniform distributions over In. The criteria for termina-
tion were met if the last k = 2 function evaluations of the iteration were below a threshold ε =
10−8 (j^F tð^sÞ ^F t1ð^sÞj < ε) with ^s 2 In being the propensity vector. The maximal number
of iterations for the heart field model and cell cycle model was 100 since a typical conver-
gence was achieved after 20–30 function evaluations.
• numerically solving
In ∍^s? : ^Fð^sÞ ¼ ^s
ð9Þ
using the nleqslv R-package [30]. More specifically, we used the Broyden secant method [31]
which is a heuristic for the Newton method. The global strategy uses the dbldog argument
which is a the trust region method using a double dogleg method [32].
Both methods yielded similar results up to solver tolerance. Graphics were generated with R
[33].
Fig 2. Parametric fixed points. An example of parametric dependency of fixed points is shown. Table 1 shows the BNE.
doi:10.1371/journal.pone.0131832.g002
Table 1. Boolean Network Extension. For a given parameter ^A, the BNE converges towards a single fixed
point. The gene expression values ^B and ^C of the fixed point depend on this parameter.
The x-axis corresponds to the parameter ^A and the y-axis shows the value of the fixed point for the variables
^B and ^C, respectively. E.g., the fixed point for ^A ¼ 0:5 is (^B ¼ 0:6; ^C ¼ 0:8).
Boolean Network
Boolean Network Extension
A
constant parameter
^A
constant parameter
B(t+1)
A_¬C(t)
^Bðt þ 1Þ
^A þ ð1 ^AÞ ð1 ^CðtÞÞ
C(t+1)
¬A_B(t)
^Cðt þ 1Þ
ð1 ^AÞ þ ^A ^BðtÞ
doi:10.1371/journal.pone.0131832.t001
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
7 / 15
Association of BNE fixed points with putative biological phenotypes
Whether considering prototypes or actual fixed points of the extension, the actual predictive
value of the model is given by its capacity to describe and predict biological phenotypes. As
mentioned above, we expect our model to reflect the properties of a Boolean state under pertur-
bation. It would hence be expected that only stable fixed points may be of interest biologically
since stable states correspond to biological states in equilibrium. Also, in simulations, unstable
fixed points are unlikely to be found numerically. However, known Boolean fixed points may
turn out to be unstable under the extension. Thus the BNE is able to additionally characterize
the stability of the Boolean fixed points. Conversely, new stable fixed points may be found by
the BNE.
The values of the fixed points do require a scheme to determine expression levels of each
specific gene to enable comparison with measured binary biological phenotypes. In practice
this would mean that we would need a binarization scheme to extract the gene expression in
terms of a set of a priori unknown critical values. For this reason we employ a more absolute
scheme for the identification by mapping fixed points and hypothetical phenotypes which lie
nearest to each other in In, in a subset of SI In, or a fuzzy description of hypothetical pheno-
types in {0, 1}n. We call this identification nearest neighbor matching (NNM). In general, using
a distance measure results in a specific ordering of the considered elements. Typically we use
the Euclidean distance since it is widely known and has an intuitive interpretation. In order to
assess the mapping approach we also applied it in context of the cell cycle network [11] (see
Section G in S1 Supplementary Information File).
Interpretation of the values of a BNE
The BNE uses fuzzy logic product-sum transformation of the Boolean rules to compute a value
for each variable. The main property of the extension is that it inherits the interaction patterns
of the BN. The defined operations corresponding to the Boolean operations are also t–norms
and corresponding associated co-norms, which ensure that the formalism is a consistent fuzzy
logic [34, 35]. Furthermore, the extension is conducted in such a way that the transition of the
DNFPS to the Boolean limit is differentiable.
We wish to emphasize that the assumption we make is that the approximation also yields
plausible results for larger perturbations over the entire In and that the intermediate values will
be in a monotonic relation to measurements of gene expression, i.e, the larger the value xi the
larger the expression of the corresponding gene product.
The assertions of the BNE values are in no way absolute but do reflect the corresponding
expression of genes in a relational way, i.e., place the corresponding expressions on an ordinal
scale. We can state, a gene has a “higher” or “lower” expression values when comparing two
values. This however does not give any conclusive answer to whether a gene is expressed or
not. The values only reveal an ordering of gene expressions. From the Boolean case we expect
that 0 corresponds to certainly unexpressed and 1 to certainly expressed. An actual quantifica-
tion for values in between 0 and 1 may hence only be carried with some a priori knowledge,
i.e., by comparison with actual measurements.
As the BNE attributes numbers to genes, it might also be natural to interpret those as con-
centrations. An example would be to associate the outputs of the BNE as the gene transcript
products over time. However, it is very unlikely that, what in effect is a system of kinetic reac-
tions may be described without the addition of any kinetic parameters. Such attempts have
been carried out in the past and do require additional parameters to describe the dynamics of
the underlying chemical molecules [20, 21]. Since the values are neither concentrations nor
probabilities we call them propensities for gene expression.
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
8 / 15
Results
Previous results for early cardiac development
Herrmann et al. [16] analyzed the gene regulatory network of early cardiac development by use
of a BN and found a correspondence between biological measurements and mathematically
simulated attractors. The model simulates the interaction between intracellular genes (Bmp2,
canonical Wnt, Dkk1, Fgf8, Foxc1.2, GATAs, Mesp1, Isl1, Nkx2.5, Tbx1, Tbx5) and extracellu-
lar factors (exogenous Bmp2 and canonical Wnt) that form gradients (Table 2).
When considering all possible binary initial values, two fixed points were reached in 99% of
the cases. The gene expression values corresponding to these two fixed points are similar to the
gene expression in the FHF and SHF that was extracted from literature. In 49% of the initial
values, the network converges to attractors corresponding to the FHF (49% of cases) and in
50% to the SHF. In the remaining 1% the simulation converged towards a fixed point with no
activated cardiac genes. This was thought to correspond to a biological phenotype where no
heart field is formed, like if canonical Wnt signaling is not activated during development. The
BN fixed point that resembles the FHF is characterized by the expression of the FHF specific
genes Bmp2, GATAs, Nkx2.5, and Tbx5 to be active. Accordingly, the fixed point for the SHF
shows activation of the SHF genes, Isl1, Foxc1/2, Tbx1 and Fgf8. In the following we name the
phenotypes predicted by the Boolean model FHF_BOOL and SHF_BOOL, respectively. We
apply the BNE to this cardiac development model in order to investigate new phenotypes that
are outside of the Boolean pradigm.
Biological phenotypes are described in terms of present or absent gene expression. In our
case we wanted to compare the gene expression of the four genes Isl1, Nkx2.5, Tbx1, and Tbx5
for FHF and SHF differentiation to the single cell RT-PCR analysis of Gessert and Kühl [19].
The phenotypes previously identified are given in Fig 3. In the following we name these pheno-
types as SHF1, SHF2, SHF3 and SHF4, as these are determined as second heart field
Table 2. Genes, proteins and model variables to BN model of cardiac development. The first column
shows the variables used in the BN model. The second column, function, describes the type and location of
the expressed protein or the purpose of the variable in the BN.
Variable (Gene/Protein)
Function
Intracellular factors
Bmp2 (Bmp2)
Signaling factor
canWnt (canonical Wnt)
canonical Wnt signaling
Dkk1 (Dkk1)
Signaling factor
Fgf8 (Fgf8)
SHF transcription factor
Foxc1.2 (Foxc1, Foxc2)
SHF transcription factor
GATAs (GATA4, GATA5, GATA6)
transcription factor, cardiogenic mesoderm
Isl1 (Isl1)
SHF transcription factor
Mesp1 (Mesp1, Mesp2)
transcription factor, early cardiogenic mesoderm development
Nkx2.5 (Nkx2.5)
transcription factor, cardiogenic mesoderm
Tbx1 (Tbx1)
SHF transcription factor
Tbx5 (Tbx5)
FHF transcription factor
Extracellular factors
exogen_Bmp2_I
Bmp2 derived from neighboring tissue
exogen_Bmp2_II
Time delay of Bmp2 derived from neighboring tissue
exogen_CanWnt_I
Canonical Wnt derived from neighboring tissue
exogen_CanWnt_II
Time delay of canonical Wnt derived from neighboring tissue
doi:10.1371/journal.pone.0131832.t002
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
9 / 15
phenotypes by the expression of the SHF marker gene Isl1, and the phenotypes representing
the first heart field FHF5, FHF6, FHF7.
Simulation results for the BNE
Given the continuous model of early cardiac development we computed fixed points for 104
different initial values of the genes. The values for each gene were drawn from a uniform distri-
bution U(0,1). We additionally performed simulations that were aimed at initial inactivation
(value 0) of all genes with the exogen_canWnt_I as controlling parameter in order to examine
its influence to the formation of the phenotypes. As the cardiac model is parametrized by the
exogen_canWnt_I parameter, we ordered the identified fixed points accordingly. The linear
increase of the propensity of the exogen_canWnt_I causes a continuous transition in propen-
sity for the remaining genes (see Fig B in S1 Supplementary Information File).
Prediction of additional phenotypes for FHF and SHF
The structure provided by the BN is sufficient enough to allow a prediction of previously
unknown biological phenotypes through the extension.
In order to characterize the phenotypes in the BNE we compared all 11 genes that are effec-
tively modeled by the BN (Bmp2, canonical Wnt, Dkk1, Fgf8, Foxc1.2, GATAs, Mesp1, Isl1,
Nkx2.5, Tbx1, Tbx5). This allows prediction of gene expression propensity of genes for which
no expression information is available. In order to evaluate our results we computed the dis-
tances between all hypothetical phenotypes and the continuous fixed points for each parameter
value of exogen_canWnt_I in the simulation. The 11 genes give rise to 211 = 2048 different
qualitative expression patterns that are compared and the phenotype with the minimal distance
Fig 3. Schematic drawing of cardiac tissue in Xenopus laevis at stage 24—Expression of genes in heart fields (left) and RT–PCR analysis of
selected genes (right). Panels adapted from Gessert and Kühl [19]. The left panel shows the genes expressed in different domains of the first heart field
(FHF) and second heart field (SHF). The SHF is shown at the top and the FHF is shown at the bottom. Common genes expressed in all regions of the FHF
and SHF, respectively, are shown on the left. Genes expressed in particular domains are shown on the right. Colors indicate different domains and
corresponding expressed genes. The right figure shows the results of single cell RT–PCR analysis of gene expression for the four genes Nkx2.5, Isl1, Tbx1,
and Tbx5. Values (0 and 1) and colors red/green represent inactive or active genes. The panel shows the gene expression of different single cell samples
(numbered and named at the bottom). FHF and SHF are distinguished by the expression of the Isl1.
doi:10.1371/journal.pone.0131832.g003
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
10 / 15
to a fixed point is then considered to be the phenotype of this fixed point. In order to distin-
guish the hypothetical phenotypes we use the naming scheme “PH-X” where X is the decimal
number encoding the binary expression pattern of the corresponding phenotype.
Fig 4 shows the 7 nearest hypothetical phenotypes that are mapped to the computed fixed
points of the BNE. For the genes Isl1, Nkx2.5, Tbx1, and Tbx5 we see the same expression pro-
pensity pattern that is referenced in the RT-PCR analysis. The phenotypes FHF6 and SHF4
also correspond to the fixed points of the BN model FHF_BOOL and SHF_BOOL, respectively.
The NO_CARDIAC fixed point of the BNE also corresponds to the “no-cardiac” fixed point of
the BN. For the remaining phenotypes BNE predicts expression pattern for the FHF7 via PH-
1060 and additionally three new phenotypes. These phenotypes, PH-1076, PH-564, and PH-
692, correspond to the SHF1 based on the four genes measured by RT-PCR [19], however, the
data shows that the SHF1 phenotype may have different gene expression propensities for the
genes Bmp2, canonical Wnt, and Fgf8 (Fig 4).
Stability of fixed points of the BN
The Boolean phenotypes predicted by the BN model correspond to the FHF6 and SHF4 pheno-
types (Figs 3 and 4). In the BNE these phenotypes correspond to values close to 0 and 1 of the
exogen_canWnt_I expression propensity. Consistently, our extension can predict the same
phenotypes of the BN model and it shows that under the perturbation of exogen_canWnt_I
Fig 4. Phenotypes predicted by the BNE. The phenotype profile used for the mapping is based on the 11 genes present in both the Boolean model and the
Xenopus analysis. The figure in the left panel shows the distance curves for the nearest phenotypes and fixed points. The x-axis denotes the values of the
parameter exogen_canWnt_I and the phenotypes to which the fixed points were mapped. The y-axis shows the actual distance. The phenotypes are ordered
by increasing exogen_canWnt_I expression propensity (right panel). Activated genes are shown in green and deactivated genes are shown in red. The
framed box shows the gene expression propensity pattern for the four genes Isl1, Nkx2.5, Tbx1, and Tbx5 that corresponds to the the RT–PCR phenotypes
of the FHF and SHF from the Fig 3. The FHF_BOOL and SHF_BOOL phenotypes correspond to the phenotypes found in the Boolean model [16]. The SHF1
phenotype is split in three sub-phenotypes PH-1076, PH-564, and PH-692 that differ by the gene expression propensity of canWnt, Bmp2, and Fgf8. The
expression of the Fgf8 gene was not reported in Xenopus. Its activation pattern is a prediction of the BNE.
doi:10.1371/journal.pone.0131832.g004
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
11 / 15
parameter the results remain close to the phenotypes found by the BN model. The 1% attractor,
which was assumed not to correspond to any of the heart fields, is also a fixed point of the
BNE. In our simulation we additionally analyzed the BN fixed points for stability. In particular,
the 1% attractor reported in the Boolean Network model. Any perturbations added to this
fixed point resulted in the fixed point corresponding to FHF4 for the given value of exogen_-
canWnt_I. This fixed point is thus unstable in the BNE.
Discussion
We extended the Boolean network model of early cardiac development and identified the bio-
logical phenotypes that were previously predicted by the Boolean model as well as additional
biological phenotypes that represent a more detailed differentiation of the FHF and SHF in
terms of gene expression propensities. These additional phenotypes were confirmed by experi-
ments in Xenopus laevis[19].
There are several advantages of the proposed method. Essentially, it helps to characterize
the gene expression propensity of phenotypes from the structure given by the BN alone. It cor-
responds to the BN model in case of binary inputs. The complexity of the extension can be
seen as an intermediate representation between the BN and ODE models. It can cope with con-
tinuous values but does not need additional kinematic parameters that are required, e.g., in an
ODE model for concentration levels. Considering the possibility theory approach [36], the
BNE does not use fuzzy sets and thus is not representable as a possibility. In our case, the fuzzy
logic approach is used to extend the Boolean function on the intervals [0, 1].
Different approaches exist that transform a BN into a system of ordinary differential equa-
tions. Mendoza and Xenarios [21] partition the genes into activating and inhibiting subsets.
Each subset is postulated to activate or inactivate a considered gene in a sigmoid manner add-
ing the corresponding terms to the ODE vector field. They do not directly transform the logical
rules of the BN, but rather construct the rules according to the activator and inhibitor subsets.
They identify fixed points in the system by perturbing the Boolean attractors. By focusing on
activation and inhibition of the genes they limit the BN to only a subset of possible Boolean
rules for state transition. Conversely, the approach of Wittmann et al. [20] directly transforms
the rules of a BN into continuous functions. They include production and decay rates into the
rules, thereby introducing additional parameters into the model. Transformation of the contin-
uous input variables to continuous switch-like values is done with Hill functions [37–39]. Both
approaches have in common that they require additional knowledge either from biological
experiments or expert opinion to determine the values for the different parameters.
The state space of BN is discrete and finite. The attractors can be exactly determined by sim-
ulating the network in an exhaustive manner. In this respect, the BNE has the same limitation
as an ODE model. In general, the complete exploration of the search space for identification of
fixed points is infeasible and we cannot determine the number of fixed points in the system a
priori. Fixed points of the model can only be found numerically by sweeping through the
search space.
The BNE behaviour resembles that of the Boolean model for small perturbations in the
Boolean input. When the Boolean input is perturbed and the functions are evaluated for a sin-
gle step, the resulting values of the BNE deviate slightly from the values of the successor state of
the corresponding BN. The BNE functions reflect the Boolean rules of the Boolean Network
which regulate the expression of genes. The BNE models the influence of genes on other genes
in a continuous manner. The resulting values of BNE can be seen as propensity for gene
expression. As genes interact with each other via the concentration of gene products, a propen-
sity is similar to concentration levels.
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
12 / 15
Similarly to the fixed points of the BN, we relate the resulting fixed points to biological phe-
notypes. We use a distance based approach to map fixed points to phenotypes. In order to
compare the fixed points we compute Euclidean distances to the biological phenotypes. The
hypothetical phenotype which has the shortest distance to a fixed point corresponds to the phe-
notype of the fixed point. In case of perfect agreement between the fixed point and the pheno-
type the distance is 0. In order to find the best corresponding phenotype to a fixed point it is
necessary to exhaustively test the set of all hypothetical phenotypes.
Since the values of genes of the fixed points represent only gene expression propensities,
one could choose a threshold for which a certain gene propensity becomes either an activation
or inactivation of a gene. However, this requires additional knowledge about the property of
the gene. Usually, the threshold is not easily determined and the choice may be arbitrary. We
avoid choosing a threshold by using the proposed distance based approach.
The fixed points of the BNE for the cardiac development are parametrized by the exogen_-
canWnt_I parameter. This parameter influences all core genes of the model except for exogenous
Bmp2 parameters. This means that the fixed points of the cardiac model are not just single points,
but the values of the fixed point are continuous curves that depend directly on the gene expres-
sion propensity of the exogenous canonical Wnt. Further, the cardiac model directly encodes the
exogen_Bmp2_I as a constant in the model. If we allow interval values for the exogen_Bmp2_I, it
would directly influence the interaction of the core genes. This would result in fixed points that
depend on two independent parameters and form a plane of fixed points. Here, however, we
want to remain faithful to the original Boolean model for cardiac development where the exo-
gen_Bmp2_I parameter is needed to be “on” in order to start the FHF and SHF formation [40].
The additional phenotypes, that are described by the fixed points of the BNE, are not found
in the BN due to its discrete nature. However, these phenotypes are found to be biologically rel-
evant to the early cardiac development. In general, the examination of BN attractors in pertur-
bative manner with the BNE makes it possible to further characterize any Boolean model. The
attractors of the cardiac development BN model are per definition fixed points in the BNE. In
case of the 1% attractor from the Boolean Model, we found that this fixed point is unstable.
Any perturbation of this fixed point leads to the FHF phenotype of the BN.
In our approach we use the nearest neighbor method to map fixed points to phenotypes.
We do so by only considering a subset of genes that are know from the literature. However,
once we have mapped the phenotypes, we can explore genes for which no information is avail-
able. The gene expression propensity of the additional genes is given by the fixed points of the
simulated model. We thus can predict what a phenotype would look like by inspecting the
complete set of genes for the nearest hypothetical phenotype as has been shown in the simula-
tion of the BNE for the 11 core genes for early cardiac development.
Supporting Information
S1 Supplementary Information File. Detailed description of the cardiac and cell cycle BN
and BNE. Additional information regarding parametric dependency and biological relevance
for the subset of modelled genes, simulations with varying exogen_Bmp2_I parameter, and
min–max operator.
(PDF)
Acknowledgments
This work was funded in part by the German federal ministry of education and research
(BMBF) within the framework GERONTOSYS II (Forschungskern SyStaR, Project ID
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
13 / 15
0315894A to MK and HAK), European Community’s Seventh Framework Programme (FP7/
2007-2013) under grant agreement n°602783 (to HAK), and the International Graduate School
in Molecular Medicine at Ulm University (GSC270).
Author Contributions
Conceived and designed the experiments: HAK MK GP. Performed the experiments: MG AB
JES. Analyzed the data: JMK JES MK. Contributed reagents/materials/analysis tools: HAK.
Wrote the paper: MG AB JES AG MK HAK.
References
1.
Davidson EH. The regulatory genome: gene regulatory networks in development and evolution. Aca-
demic Press; 2006.
2.
Kestler HA, Wawra C, Kracher B, Kühl M. Network modeling of signal transduction: establishing the
global view. Bioessays. 2008; 30(11–12):1110–1125. doi: 10.1002/bies.20834
3.
Kestler HA, Kühl M. From individual Wnt pathways towards a Wnt signalling network. Phil Trans R Soc
B. 2008; 363:1333–1347. doi: 10.1098/rstb.2007.2251
4.
Shmulevich I, Dougherty ER. Genomic Signal Processing. Princeton University Press; 2007.
5.
Naldi A, Monteiro PT, Müssel C, the Consortium for Logical Models and Tools, Kestler HA, Thieffry D,
et al. Cooperative development of logical modelling standards and tools with CoLoMoTo. Bioinformat-
ics. 2015;. doi: 10.1093/bioinformatics/btv013
6.
de Jong H. Modeling and Simulation of Genetic Regulatory Systems: A Literature Review. Journal of
Computational Biology. 2002; 9(1):67–103. doi: 10.1089/10665270252833208
7.
Bornholdt S. Less Is More in Modeling Large Genetic Networks. Science. 2005; 310:449–451. doi: 10.
1126/science.1119959
8.
Hopfensitz M, Müssel C, Maucher M, Kestler HA. Attractors in Boolean networks—a tutorial. Computa-
tional Statistics. 2013; 28(1):19–36. doi: 10.1007/s00180-012-0324-2
9.
Han S, Wong RKW, Lee TCM, Shen L, Li SR, Fan X. A Full Bayesian Approach for Boolean Genetic
Network Inference. PLoS ONE. 2014; 9:e115806. doi: 10.1371/journal.pone.0115806
10.
Dümcke S, Bräuer J, Anchang B, Spang R, Beerenwinkel N, Tresch A. Exact likelihood computation in
Boolean networks with probabilistic time delays, and its application in signal network reconstruction.
Bioinformatics. 2014; 30:414–419. doi: 10.1093/bioinformatics/btt696
11.
Fauré A, Naldi A, Chaouiya C, Thieffry D. Dynamical analysis of a generic Boolean model for the control
of the mammalian cell cycle. Bioinformatics. 2006; 22(14):e124–e131. doi: 10.1093/bioinformatics/
btl210
12.
Li S, Assmann SM, Albert R. Predicting Essential Components of Signal Transduction Networks: A
Dynamic Model of Guard Cell Abscisic Acid Signaling. PLoS Biol. 2006; 4(10):e312. doi: 10.1371/
journal.pbio.0040312
13.
Sridharan S, Layek R, Datta A, Venkatraj J. Boolean modeling and fault diagnosis in oxidative stress
response. BMC Genomics. 2012; 13(Suppl 6):S4. doi: 10.1186/1471-2164-13-S6-S4
14.
Berestovsky N, Nakhleh L. An Evaluation of Methods for Inferring Boolean Networks from Time-Series
Data. PLoS One. 2013; 8(6):e66031. doi: 10.1371/journal.pone.0066031
15.
Lescroart F, Chabab S, Lin X, Rulands S, Paulissen C, Rodolosse A, et al. Early lineage restriction in
temporally distinct populations of Mesp1 progenitors during mammalian heart development. Nat Cell
Biol. 2014; 16(9):829–840. doi: 10.1038/ncb3024
16.
Herrmann F, Groß A, Zhou D, Kestler HA, Kühl M. A Boolean Model of the Cardiac Gene Regulatory
Network Determining First and Second Heart Field Identity. PLoS One. 2012; 7(10):e46798. doi: 10.
1371/journal.pone.0046798
17.
Abu-Issa R, Kirby ML. Heart Field: From Mesoderm to Heart Tube. Annu Rev Cell Dev Biol. 2007;
23:45–68. doi: 10.1146/annurev.cellbio.23.090506.123331
18.
Gessert S, Kühl M. The Multiple Phases and Faces of Wnt Signaling During Cardiac Differentiation and
Development. Circ Res. 2010; 107:186–199. doi: 10.1161/CIRCRESAHA.110.221531
19.
Gessert S, Kühl M. Comparative gene expression analysis and fate mapping studies suggest an early
segregation of cardiogenic lineages in Xenopus laevis. Dev Biol. 2009; 334:395–408. doi: 10.1016/j.
ydbio.2009.07.037
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
14 / 15
20.
Wittmann DM, Krumsiek J, Saez-Rodriguez J, Lauffenburger DA, Klamt S, Theis FJ. Transforming
Boolean models to continuous models: methodology and application to T-cell receptor signaling. BMC
Syst Biol. 2009; 3(98):1–21.
21.
Mendoza L, Xenarios I. A method for the generation of standardized qualitative dynamical systems of
regulatory networks. Theor Biol Med Model. 2006; 3(13):1–18.
22.
Morris MK, Saez-Rodriguez J, Sorger PK, Lauffenburger DA. Logic-Based Models for the Analysis of
Cell Signaling Networks. Biochemistry. 2010; 49(15):3216–3224. doi: 10.1021/bi902202q
23.
MacNamara A, Terfve C, Henriques D, Bernabé BP, Saez-Rodriguez J. State–time spectrum of signal
transduction logic models. Phys Biol. 2012; 9:045003. doi: 10.1088/1478-3975/9/4/045003
24.
Zadeh LA. Fuzzy Sets. Information and Control. 1965; 8(3):338–353. doi: 10.1016/S0019-9958(65)
90241-X
25.
Kauffman SA. Metabolic stability and epigenesis in randomly constructed genetic nets. J Theor Biol.
1969; 22(3):437–467. doi: 10.1016/0022-5193(69)90015-0
26.
Kauffman SA. The Origins of Order: Self-Organization and Selection in Evolution. Oxford University
Press; 1993.
27.
Dubois DJ. Fuzzy Sets and Systems: Theory and Applications. Academic Press; 1980.
28.
Yao J, Yao Y, Kreinovich V, da Silva PP, Starks SA, Xiang G, et al. Towards More Adequate Represen-
tation of Uncertainty: From Intervals to Set Intervals, with the Possible Addition of Probabilities and Cer-
tainty Degrees. In: Wang J, editor. 2008 IEEE International Conference on Fuzzy Systems. vol. 16.
IEEE; 2008. p. 983–990.
29.
Müssel C, Hopfensitz M, Kestler HA. BoolNet—an R package for generation, reconstruction and analy-
sis of Boolean networks. Bioinformatics. 2010; 26:1378–1380. doi: 10.1093/bioinformatics/btq124
30.
Hasselman B. nleqslv: Solve systems of non linear equations; 2014. R package version 2.1.1. Available
from: http://CRAN.R-project.org/package = nleqslv.
31.
Broyden CG. A Class of Methods for Solving Nonlinear Simultaneous Equations. Math Comput. 1965;
19:577–593. doi: 10.1090/S0025-5718-1965-0198670-6
32.
Powell MJD. A hybrid method for nonlinear equations. In: Rabinowitz P, editor. Numerical methods for
nonlinear algebraic equations. Newark: Gordon and Breach Science Publishers; 1970. p. 87–114.
33.
R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria; 2014. Avail-
able from: http://www.R-project.org/.
34.
Klement EP. Construction of Fuzzy σ-Algebras Using Triangular Norms. Journal of Mathematical Anal-
ysis and Applications. 1982; 85:543–565. doi: 10.1016/0022-247X(82)90015-4
35.
Alsina C, Trillas E, Valverde L. On Some Logical Connectives for Fuzzy Sets Theory. Journal of Mathe-
matical Analysis and Applications. 1983; 93:15–23. doi: 10.1016/0022-247X(83)90216-0
36.
Zadeh LA. Fuzzy Sets as a Basis For A Theory of Possibility. Fuzzy Sets and Systems. 1978; 1:3–28.
doi: 10.1016/0165-0114(78)90029-5
37.
Wittmann DM, Blöchl F, Trümbach D, Wurst W, Prakash N. Spatial Analysis of Expression Patterns
Predicts Genetic Interactions at the Mid-Hindbrain Boundary. PLoS Comput Biol. 2009; 5(11):
e1000569. doi: 10.1371/journal.pcbi.1000569
38.
Franke R, Theis FJ, Klamt S. From Binary to Multivalued to Continuous Models: The lac Operon as a
Case Study. J Integr Bioinform. 2010; 7(1):151.
39.
Cornelius SP, Kath WL, Motter AE. Realistic control of network dynamics. Nat Commun. 2013; 4:1942.
doi: 10.1038/ncomms2939
40.
Ma L, Lu MF, Schwartz RJ, Martin JF. Bmp2 is essential for cardiac cushion epithelial-mesenchymal
transition and myocardial patterning. Development. 2005; 132:5601–5611. doi: 10.1242/dev.02156
A Boolean Network Model Incorporating Uncertainty
PLOS ONE | DOI:10.1371/journal.pone.0131832
July 24, 2015
15 / 15
|
26207376
|
Tbx1 = ( Foxc1.2 )
Fgf8 = ( ( Tbx1 ) AND NOT ( Mesp1 ) ) OR ( ( Foxc1.2 ) AND NOT ( Mesp1 ) )
Tbx5 = ( ( ( ( Tbx5 ) AND NOT ( Tbx1 ) ) AND NOT ( Dkk1 AND ( ( ( NOT Mesp1 AND NOT Tbx5 ) ) ) ) ) AND NOT ( canWnt ) ) OR ( ( ( ( Nkx2.5 ) AND NOT ( Tbx1 ) ) AND NOT ( Dkk1 AND ( ( ( NOT Mesp1 AND NOT Tbx5 ) ) ) ) ) AND NOT ( canWnt ) ) OR ( ( ( ( Mesp1 ) AND NOT ( Tbx1 ) ) AND NOT ( Dkk1 AND ( ( ( NOT Mesp1 AND NOT Tbx5 ) ) ) ) ) AND NOT ( canWnt ) )
exogen_CanWnt_II = ( exogen_CanWnt_I )
Nkx2.5 = ( Tbx1 ) OR ( GATAs AND ( ( ( Bmp2 ) ) ) ) OR ( Tbx5 ) OR ( Isl1 AND ( ( ( GATAs ) ) ) ) OR ( Mesp1 AND ( ( ( Dkk1 ) ) ) )
Foxc1.2 = ( canWnt AND ( ( ( exogen_CanWnt_II ) ) ) )
Bmp2 = ( ( exogen_BMP2_II ) AND NOT ( canWnt ) )
GATAs = ( Tbx5 ) OR ( Nkx2.5 ) OR ( Mesp1 )
Mesp1 = ( ( canWnt ) AND NOT ( exogen_BMP2_II ) )
canWnt = ( exogen_CanWnt_II )
exogen_CanWnt_I = ( exogen_CanWnt_I )
Isl1 = ( Tbx1 ) OR ( Mesp1 ) OR ( Fgf8 ) OR ( canWnt AND ( ( ( exogen_CanWnt_II ) ) ) )
Dkk1 = ( Mesp1 ) OR ( ( canWnt ) AND NOT ( exogen_BMP2_II ) )
exogen_BMP2_II = ( exogen_BMP2_I )
|
RESEARCH ARTICLE
An Extended, Boolean Model of the Septation
Initiation Network in S. pombe Provides
Insights into Its Regulation
Anastasia Chasapi1☯, Paulina Wachowicz2☯, Anne Niknejad1, Philippe Collin2¤,
Andrea Krapp2, Elena Cano2, Viesturs Simanis2*, Ioannis Xenarios1,3*
1 Vital-IT Group, Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland, 2 Cell cycle control
laboratory, Ecole Polytechnique Fédérale de Lausanne (EPFL), SV-ISREC, Lausanne, Switzerland,
3 Swiss-Prot Group, Swiss Institute of Bioinformatics (SIB), Geneva, Switzerland
☯These authors contributed equally to this work.
¤ Current address: Horizon Discovery Group, Campbridge Research Park, Cambridge, United Kingdom
* viesturs.simanis@epfl.ch (VS); ioannis.xenarios@isb-sib.ch (IX)
Abstract
Cytokinesis in fission yeast is controlled by the Septation Initiation Network (SIN), a protein
kinase signaling network using the spindle pole body as scaffold. In order to describe
the qualitative behavior of the system and predict unknown mutant behaviors we decided
to adopt a Boolean modeling approach. In this paper, we report the construction of an
extended, Boolean model of the SIN, comprising most SIN components and regulators as
individual, experimentally testable nodes. The model uses CDK activity levels as control
nodes for the simulation of SIN related events in different stages of the cell cycle. The
model was optimized using single knock-out experiments of known phenotypic effect as a
training set, and was able to correctly predict a double knock-out test set. Moreover, the
model has made in silico predictions that have been validated in vivo, providing new insights
into the regulation and hierarchical organization of the SIN.
Introduction
Schizosaccharomyces pombe, commonly referred as fission yeast, has long been used as a model
organism for the study of conserved, essential functions in the eukaryotic cell. It has proved
highly informative in the study of the cell cycle, particularly the control of the G2/M transition.
Like many somatic higher eukaryotic cells, it divides by binary fission. Cytokinesis in fission
yeast is controlled by the Septation Initiation Network (SIN), a protein kinase signaling net-
work, which uses the spindle pole body (SPB; the functional counterpart of the centrosome in
yeast), as a scaffold from which to initiate signaling. Elements of the SIN signaling architecture
have been conserved throughout evolution. In Saccharomyces cerevisiae the corresponding
pathway is known as the mitotic exit network (MEN), and controls both cytokinesis and
mitotic exit. In higher eukaryotes the equivalent signaling network is the hippo pathway, which
regulates cell growth and proliferation [1,2].
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
1 / 22
OPEN ACCESS
Citation: Chasapi A, Wachowicz P, Niknejad A,
Collin P, Krapp A, Cano E, et al. (2015) An Extended,
Boolean Model of the Septation Initiation Network in
S. pombe Provides Insights into Its Regulation. PLoS
ONE 10(8): e0134214. doi:10.1371/journal.
pone.0134214
Editor: Takashi Toda, Cancer Research UK London
Research Institute, UNITED KINGDOM
Received: July 3, 2015
Accepted: July 9, 2015
Published: August 5, 2015
Copyright: © 2015 Chasapi et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Funding: This work was funded by a SINERGIA
grant from the Swiss National Science Foundation
(CRSII3_132392), a Swiss National Science
Foundation grant to VS (31003A_138176) and PC
(109454) and State Secretariat for Research and
Innovation (SEFRI) for IX and AN. PC contributed to
the study while being affiliated to VS's research
group, before his association with his current
affiliation with the Horizon Discovery Group. The
Horizon Discovery Group had no funding role, and no
The SIN comprises a group of protein kinases and their regulators that induce cytokinesis
when CDK activity drops in anaphase [3–5]. Signaling failure results in multinucleated cells, as
cytokinesis fails while growth and the nuclear cycle continue [6], which is referred to as the
SIN phenotype. Failure to turn off SIN signaling produces multiseptated cells that remain
uncleaved and contain one or two nuclei [7]. Ppc89p, Cdc11p and Sid4p form the scaffold
upon which signaling proteins are assembled at the SPB [8–11]. SIN signaling requires the
action of three kinase complexes. The association properties of SIN proteins with the SPB differ
in early and late mitosis (see [12], and references for each protein cited below). The kinase
Cdc7p associates with the signaling GTPase Spg1p [13,14], Sid1p associates with its regulatory
subunit Cdc14p [4,15] and the kinase Sid2p associates with its regulator Mob1p [16–18]. Asso-
ciation of the SIN kinase modules with the SPB during mitosis is considered to indicate that
the kinase in question is active (reviewed by [19,20]). The nucleotide status of Spg1p is regu-
lated by a bipartite GAP, composed of a catalytic subunit (Cdc16p), which interacts with Spg1p
in the context of a scaffold, Byr4p [21,22]. Etd1p regulates the nucleotide status of Spg1p, per-
haps by modulating Rho1p signaling [23–26]. Plo1p acts upstream of the SIN [27,28] and coor-
dinates SIN activity with other mitotic events. The SIN controls many aspects of cytokinesis
including the assembly of the contractile ring and synthesis of the division septum [29].
Our goal is to describe the qualitative behavior of the system, investigate the role of each
SIN regulator and potentially predict unknown mutant behaviors. Towards this end we
adopted a Boolean modeling approach. The choice of qualitative modeling was based on their
suitability to simulate systems with restricted kinetic data, as well as their computational effi-
ciency, that permits large numbers of in silico experiments even in networks with hundreds of
nodes.
Computational models find their origins in engineering science, and have proved to be useful
tools with which to analyze complex biological systems (for example [30–32]). The different
types of modeling techniques can vary from qualitative Boolean models, to quantitative kinetic-
based models; which of them is chosen depends on the type and amount of knowledge and
experimental data available for the specific system, as well as the size of the network [33,34].
The cell cycles of fission and budding yeast have long been popular fields of research and
several modeling strategies have been employed to understand them [30,35–40]. Models
focused on the fission yeast SIN have already been generated by Csikasz-Nagy et al. (2007) and
Bajpai et al. (2013) [41,42]. In the study by Csikasz-Nagy et al. the timing of septation in wild
type and mutant cells was described using a minimal, continuous model. The SIN components
were treated as two groups, the “Top of SIN” and “Bottom of SIN”, with Sid1p localization to
the SPB being the pivotal event that differentiates the two groups [42]. In the subsequent
model [41], the asymmetric distribution of molecules at the SPBs was analyzed using a simple,
non-linear model of two antagonistic molecules. The model was also extended to incorporate
key regulators of the SIN [41].
In this work, we present an extended, Boolean model of the SIN, comprising most known
SIN components and regulators as individual, experimentally testable nodes. The Boolean
framework allows us to perform in silico knock-out and “constant activation” experiments for
every combination of molecules present in the model, and to assess phenotypic predictions
that could be subsequently validated experimentally. Our model provided useful insights for
several aspects of SIN regulation such as the role of Fin1p, the inhibitory function of Nuc2p in
interphase, as well as an in silico, counter-intuitive, double mutant phenotypic prediction. The
model predicted that Sid4p mutant cells would septate if they express Cdc7p in high levels. The
prediction has been experimentally confirmed. This work serves as a good example of the use
of qualitative modeling in hypotheses generation and prediction of experimental outcomes in
otherwise complicated and long experiments.
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
2 / 22
input into the design or execution of the study. The
funders had no role in study design, data collection
and analysis, decision to publish, or preparation of
the manuscript.
Competing Interests: The authors declare that they
have no conflicts of interest.
Results
Model construction through expert biocuration
An overview of the workflow used for the model construction, optimization and use is pre-
sented in Fig 1. For the gene regulatory network construction of the SIN we chose an expert
biocuration approach [43,44], taking advantage of the long-term expertise in the Swiss-Prot
group. Experimentally determined interactions specific to the SIN, were retrieved, structured,
curated and annotated from the literature and from available knowledge databases (for exam-
ple Pubmed, iHOP, UniProtKB/Swiss-Prot, ChEBI). To generate the model, we started by add-
ing the main SIN signaling regulators such as the GTPase Spg1p, its effector kinase Cdc7p and
the GAP Byr4p and Cdc16p [13,21,45]. We then added the SPB scaffold for the SIN, which is
comprised of Ppc89p, Sid4p and Cdc11p [10,45]. Subsequently, additional regulators were
added to this core unit, to complete a first working model. The collected knowledge was stored
in a structure formed of pairwise interactions and regulations that include information about
participating components, the origin of publications (PMID), the evidence used to evaluate the
interaction was mentioned and a confidence assessment as an evidence tag from the biocurator
(a full interaction table provided in S1 Table).
The constructed prior knowledge network (PKN) consists of 50 nodes (gene products, pro-
teins and complexes) and 124 directed edges (Fig 2A). The regulatory information is the result
of the curation of 67 published scientific papers (S1 table). The most recently published interac-
tion contained in this model is the inhibitory regulation of CDK and Plo1p upon Byr4p
recently published by [46].
The model interactions were classified as activations or inhibitions and they were repre-
sented in the network as a combination of Boolean functions that can include AND, OR and
NOT [40,47–51].
Qualitative model simulation
Despite the intensive study of the SIN over the past decades, there is little kinetic data for the
protein interactions described in the literature that form the basis for our model. Obtaining
such spatiotemporal data is experimentally difficult and represents one of the major challenges
in systems biology research. For the simulation of the SIN model we adopted a qualitative Bool-
ean approach, which has been successfully used in several other contexts [30,40,52–62].
In Boolean formalism, each node is characterized by an activation state that can take the val-
ues 1 for “active” or 0 for “inactive”, corresponding to the logical values TRUE and FALSE. The
activation state can refer to transcription, localization, phosphorylation or other post-transla-
tional modifications. For the construction of the SIN model we assumed that for scaffold pro-
teins “TRUE” corresponds to a state that permits the assembly of signaling complexes.
The state of each node depends on the state of the nodes regulating it, that is, the state of all
the incoming edges, and the rules that govern their interaction. The state of all the nodes at a
given moment defines the network state. The network transitions from state to state are dic-
tated by the underlying Boolean functions, until it reaches a steady state or a cyclic attractor
[48]. The possible trajectories in the state space can be represented by the state transition graph
[34,63,64].
In Boolean modeling, time is abstract and can be simulated using diverse strategies such as
in a continuous manner, with discrete updates or using probabilistic transitions. In the case of
discrete time representation, two main updating schemes can be used during model simulation;
synchronous and asynchronous update. The former assumes that all biological events in the
system have similar timescales, and all functions are updated simultaneously. In the latter, one
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
3 / 22
function is updated at each time step, which can be deterministic (deterministic asynchronous)
or randomly selected (stochastic asynchronous) [65,66]. The asynchronous behavior can be
controlled by setting additional rules for time delays and priorities [67,68]. Alternatively, all
possible transitions can be generated [33,34,67,69].
Fig 1. Model construction and optimization workflow. The Prior Knowledge Network (PKN) is constructed
after collecting relevant information from various sources, including network databases and literature. The
PKN is translated into logical functions, describing the regulatory relations among gene products. The logical
model is simulated under the preferred conditions, resulting in one or more steady states, where all logical
rules are satisfied. The model goes then through an optimization procedure, where the goal is to fit the
resulting steady states with available experimental data by altering regulatory rules. The optimization typically
includes removing outdated / low confidence links, adjusting their representation and adding new regulatory
rules. The process is iterated until the simulation fits the available data. The model can then be used as a
predictive tool, by performing in silico perturbations. Validation of the predictions can lead to discovery of
missing regulatory links that are then added to the PKN.
doi:10.1371/journal.pone.0134214.g001
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
4 / 22
Fig 2. The extended Boolean SIN model. (A) The initial, prior knowledge network, manually re-constructed from the literature. Purple nodes represent
proteins and complexes that take part in the regulation of the SIN, and pink nodes represent AND gates. Blue arrows indicate activation events and orange
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
5 / 22
Asynchronous deterministic updating was chosen for the SIN model simulation in this
study, since it assumes non-synchronous regulatory events, which is likely to reflect the in vivo
situation. However, the challenge in asynchronous update lies in interpreting the simulation
trajectories; in stochastic asynchronous simulations, the same initial state can lead to different
trajectories in the state space, due to the stochasticity of the updating scheme [34]. The simula-
tion algorithm used was based on genysis, a tool for synchronous and asynchronous modeling
of gene regulatory networks, based on reduced ordered binary decision diagrams (ROBDDs)
[69]. The algorithm identifies all steady states / attractors that can be reached, by efficiently
investigating all possible asynchronous state transitions.
The use of nodes representing CDK levels as input nodes for SIN
activity modeling
Cdc2p/CDK1 influences the SIN both positively and negatively. Active Cdc2p inhibits the SIN
early in mitosis; its inactivation is required for septum formation and to establish SIN protein
asymmetry [3,4,70–72]. Furthermore, Cdc2p and the Byr4p-Cdc16p GAP may cooperate to
prevent septation in interphase [73]. However, Cdc2p and Plo1p also collaborate positively to
ensure removal of Byr4p from the SPBs and facilitate SIN signaling in anaphase [46]. Failure to
increase CDK levels during early mitosis will block cytokinesis, since the cells do not enter
mitosis. However, constant, high CDK levels through mitosis will block cytokinesis. Thus,
CDK levels need to increase to permit entry into mitosis, after which cytokinesis will occur.
However, this will only happen once CDK activity decreases to a very low level, and cells exit
mitosis. The model must therefore accommodate these CDK-dependent regulatory events.
Towards this goal, we introduced three independent nodes for CDK, representing the CDK
levels before, during and after mitosis. CDK-L corresponds to the low CDK levels during inter-
phase; these prevent re-replication of DNA, but are insufficient for entry to mitosis [74,75],
CDK-H represents the high level of CDK activity found in early mitosis. Finally, CDK-0 repre-
sents the very low CDK activity in late mitosis as cells undergo the M-G1 transition. This
multi-node representation of CDK allows us to describe the SIN-related phenotypes corre-
sponding to several stages of the cell cycle, using the CDK nodes as inputs. For example, setting
CDK-L constantly on, indicates that we are simulating the events during interphase, while,
CDK-H on represents early mitosis and CDK-0 on represents late mitosis (Fig 2B). It should
be stressed that the 3 CDK nodes are not regulated themselves, but are rather used as control
(i.e. input) nodes for the system’s simulation. For this reason, there are no incoming regulatory
links towards the CDK nodes (CDK-L, CDK-H and CDK-0) (Fig 2B).
Model refinement and simulation results
This model configuration that uses CDK levels as control nodes for the simulation of cell cycle
events, allowed us to clearly define the expected steady states of the system and set our refine-
ment strategy. First, we attributed the PKN interactions involving CDK to the correct CDK
node. For example, an activation link from CDK-H was added towards Plo1p (Fig 2B), which
in turn will reinforce the activity of CDK in a positive feedback loop [28,76]. Following the
attribution step, the model was evaluated using a number of well characterized in silico
circles inhibition events. Other logical functions, such as AND, OR and NOT regulatory gates are also encoded in the model. SAC: spindle assembly
checkpoint, APC: anaphase-promoting complex, PP: protein phosphatase. (B) The final, optimized model, which uses a 3-node representation of CDK
activity. Nodes in green are used as switches, and they are turned on to represent different stages of the cell cycle: interphase, early mitosis and late mitosis.
Pink nodes represent AND gates. In the case of the Cdc42p regulatory link towards Byr4p, the regulatory rule can be phrased as “NOT Cdc42p activates
Byr4p”.
doi:10.1371/journal.pone.0134214.g002
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
6 / 22
perturbations whose phenotypic consequences are known; knock-out of cdc11, spg1, cdc16,
byr4, and cdc7. For the evaluation, the above 5 knock-out perturbations were simulated, by set-
ting the corresponding node to 0 throughout simulation. A fixed set of nodes, with activation
states indicative of the expected phenotype was selected to score the model’s ability to correctly
reproduce the mutation outcomes. The scoring set includes sid4, cdc11, byr4-cdc16, spg1, cdc7,
sid2-mob1 and sid1-cdc14. For each in silico perturbation, the resulting steady states were eval-
uated according to the number of the scoring set nodes that had the expected activation state
(see S1 Fig for a list of the scoring set expected states).
We proceeded by refining the connections within the network. A refinement cycle consisted
of altering an edge of the network, perturbing the model and evaluating the simulation out-
come of the perturbations test set. The alterations could involve additions and deletions of reg-
ulatory edges, or modifications of the existing regulatory rules. The reasoning behind each
change of the model’s regulatory rules was based on several factors, such as the confidence
level of each interaction, coupled with information from the published literature, as well as
forming alternative logical rules of the given information to better represent the biological real-
ity of the interaction. For example, “A inhibits B” can be alternatively encoded as “NOT A acti-
vates B”, and is more suited for cases where the inhibition is not dominant. During this process
we maintained the known, required connections of the model and minimized the model’s com-
plexity by removing nodes that no longer served any regulatory role in the model. An example
of the latter is the removal of cell cycle regulatory elements such as Cdc25p, Wee1p, Slp1p and
Rum1p, to simplify the cell cycle representation by using multi-node CDK. The final, opti-
mized network is presented in Fig 2B. A full list of the edges comprising the final network,
together with the justification for the inclusion of each edge, can be found at the supplementary
material (S2 Table), as well as the model in genYsis and SBML-Qual format (S1 and S2 Files
respectively) [77].
The optimized model was used for in silico experiments in which a combination of nodes
was perturbed and the phenotypic outcome in the interphase, early mitosis and late mitosis
CDK-states were determined. A simulation of the wild type model, where no perturbation is
introduced, is presented in Fig 3.
To simulate interphase, CDK-L is set to 1, and Ppc89p is set to 1 as well, to permit “binding”
of scaffold proteins to the SPB. In interphase, the model simulation results in a steady state
where Byr4p and Cdc16p are present and able to form the GAP complex, therefore active. The
scaffold proteins Sid4p and Cdc11p are also present (therefore “active” according to our initial
assumption for scaffold molecules), but no SIN signaling occurs due to the inhibitory effect of
the Byr4p-Cdc16p GAP.
Early mitosis is simulated by setting CDK-H and Ppc89p to 1. The SIN scaffold is still
formed, as expected. Cdc16p is absent from the SPBs in early mitosis, preventing formation of
the GAP. This allows SIN signaling to initiate, and we observe that all the main components of
the SIN are active (Plo1p, Spg1p, Cdc7p, Sid2p-Mob1p), apart from Cdc14p-Sid1p, which is
inhibited by high CDK activity [4,70].
Late mitosis is represented by setting CDK-0 and Ppc89p to 1 during the simulation. There
are 2 resulting steady states of the system simulation. In one state, the SIN signaling scaffold is
present, the Byr4p-Cdc16p complex is formed, and all SIN components, except Plo1p are inac-
tive. In the other state, Byr4p-Cdc16p is not active, and all proteins of the SIN scaffold and sig-
naling including Cdc14p-Sid1p are active. Intriguingly, these resemble the asymmetric
constellation of proteins observed at the old and new SPBs in late anaphase B (see [19,20,29]
for review), with the exception of Sid2p-Mob1p, which is present on both SPBs, but only active
in one of the two states of the model. Setting GAP function to 0 abolishes the state that resem-
bles the old SPB. Though it is often assumed to be the case, there is scant evidence to support
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
7 / 22
the view that localization of SIN proteins to the SPB is a faithful readout of their in vivo activity.
There is no data addressing whether Sid2p signals from one or two SPBs in late anaphase.
Fig 3. In silico steady states of the SIN, in wild type and mutated cells. Steady states deriving from simulations performed on the final model. The boxes
on the left indicate the experiments performed, which can be knock-out (KO) or over-expression (OE). When there is more than one gene in the box, it is a
double perturbation. For each perturbation, 3 experiments were performed: interphase simulation (indicated as i), early mitosis (eM) and late mitosis (lM, with
suffixes new and old when there are 2 resulting steady states, indicative of late mitosis asymmetry). Blue boxes correspond to active proteins, white to
inactive and light blue to proteins that can be either active or inactive at the resulting steady states of the system.
doi:10.1371/journal.pone.0134214.g003
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
8 / 22
Future experiments will investigate this. A detailed heatmap showing the activation state of all
nodes of the model for all experiments presented herein can be found in the supplementary
material (S2 Fig).
Assessing experimentally validated in silico perturbations for the model
evaluation
The optimized model can describe the SIN related events during interphase, early and late
mitosis. In order to evaluate the model’s ability to describe current knowledge regarding S.
pombe mutants, we performed a series of in silico knock-out and constant activation experi-
ments mimicking those described in the literature that have an established phenotype. Fig 3
summarizes the steady states yielded after simulating interphase, early and late mitosis behav-
ior of core gene mutants. Interestingly, in all the in silico experiments we obtained steady states
where the nodes displayed, overall, the expected activation state. More specifically, cdc11
knock-out completely blocks septation. Both, byr4 knock-out and cdc16 knock-out have the
same effect, which is failure to inhibit SIN signaling, and therefore SIN triggering in interphase.
In a knock-out of either spg1 or cdc7, signaling fails, with Spg1p still getting activated in cdc7
deletion, indicating that Spg1p acts upstream of Cdc7p, as experimentally proven.
Apart from the experiments that were used as training set for the model refinement, we per-
formed double mutant experiments towards which the model had not been optimized (test
set). These experiments assess the predictive value of the model, as the in silico predictions are
in accordance with the expected results. Specifically, the double deletion of cdc11 and cdc16
simulation predicts that cells should not septate, as shown in Fig 3, with supporting evidence
from the literature [78]. A Cdc7p over-expression in an spg1 deletion mutant will septate, in
agreement with in vivo studies [13]. Moreover, Cdc7p over-expression will produce septation
in the absence of Cdc11p (Fig 4A), as confirmed by the literature [79]. In this project, setting a
node to 1 throughout the simulation has been used to simulate over-expression in silico, except
in cases where it is known that the over-expression phenotype results from an indirect effect,
such as the titration of another protein.
Other in silico experiments performed during the optimization provided us with great
insights into potential knowledge gaps regarding SIN regulation, as well as the limitations of
our model. One such example was a prediction that a byr4-null sid4-null should septate. When
this was tested in vivo, the double mutant cells did not septate. This allowed us to refine the
model, by identifying regulatory links that would permit this state to be achieved and target
them as candidates for edge deletion. Moreover, the nuc2 inhibitory links that were present in
the PKN revealed our limitation of describing events that occur at the end of septation and the
incomplete regulatory inputs to cdc16 helped us discover a potential link with fin1. The nuc2
and fin1 cases are discussed in detail below.
Does Nuc2p have a role in interphase?
Increased expression of the APC/C component nuc2 blocks septation, while incubation of
nuc2-663 at low restrictive temperature results in cutting of the cell [82,83]. Analysis of how
the SIN is reset at the end of mitosis revealed an APC/C-independent role for Nuc2p [84].
Nuc2p interferes with formation of the Cdc7p-Spg1p complex, possibly by stimulating the
GAP activity of Byr4p-Cdc16p. Since our current model does not include resetting of the SIN,
we tested whether the inhibitory link of nuc2 towards the Cdc7p-Spg1p complex should be
maintained. If it is required, then it might indicate a role for Nuc2p in regulating septation in
interphase, once the cell has completed the M-G1 transition. We therefore modeled the effect
of inactivating Nuc2p in silico upon SIN behavior in interphase. The predicted outcome when
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
9 / 22
including the Nuc2p inhibitory link was two steady states; one with inactive SIN and one with
cells that septate in interphase.
To test whether this could be the case in vivo, the strain nuc2-663 atb2-mCherry leu1-32 was
arrested in S-phase by growth in medium containing 12mM hydroxyurea (HU). After 5h at
25°C, cells were shifted to 36°C to inactivate Nuc2p, and samples were analyzed at hourly inter-
vals. Before shift to 36°C nuc2-663 (97%; N = 403 cells) and nuc2+ cells (97%; N = 480) were
mononucleate with no septum; the interphase arrest was confirmed by the presence of an inter-
phase array of microtubules (data not shown). Following shift of the cultures to 36°C, the
majority of nuc2+ cells remained in interphase for three hours, as judged by the continued pres-
ence of interphasic microtubules and the absence of a spindle (97%; N = 498, and 90%; N = 400
Fig 4. Cdc7p over-expression in a sid4 mutant will result in septation. (A) Steady states of in silico, double mutation experiments. The model predicts
that in the absence of SIN scaffold proteins (Cdc11p or Sid4p) and over-expression of Cdc7p, the cell will septate. (B) sid4-SA1 leu1-32 was transformed with
a REP1-based plasmids [80] expressing cdc7; empty vector served as a control. Cells were grown to exponential phase in EMM2 medium at 25°C containing
2mM thiamine. Expression was induced by washing with EMM2 and growth for 16h at 25°C; cells were then shifted to 36°C for 5h, fixed, and stained with
DAPI and Calcofluor as described [81]. Note that the cells carrying empty vector have become elongated and multinucleated, while 75% of cells expressing
cdc7 have one or more septa. The scale bar represents 10 μm. (C) The strain leu1::pADH1-cdc7 was grown to exponential phase in YE medium at 19°C. A
sample was taken and cells were fixed and stained with DAPI and Calcofluor. The remainder of the culture was incubated for 5h at 36°C before fixation. Note
the elevated percentage of septated cells. The scale bar represents 10 μm. (D) The indicated strains were grown to exponential phase in YE medium,
counted, and diluted to 106 ml-1. 10 μl of serial 5-fold dilutions were spotted on plates, allowed to dry and then incubated at the indicated temperature until the
wild-type control had formed colonies.
doi:10.1371/journal.pone.0134214.g004
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
10 / 22
at, 1 and 2h respectively;). The nuc2-663 cells maintained the hydroxyurea arrest less effi-
ciently, (89%; N = 319, and 86%; N = 636 at 1, and 2h, respectively). When nuc2-663 cells
entered mitosis, they arrested with a mitotic spindle (not shown). A fraction of both nuc2+ and
nuc2-663 cells septated in the first two hours, but this did not exceed 5%. In contrast, previous
studies from this lab [73] and the Hagan lab [85] showed that activation of the SIN in inter-
phase-arrested cells by incubation of the cdc16-116 mutant at 36°C produced >50% of type II
(mononucleated, septated cells; defined by Minet et al. [7]) within 100 minutes. The levels of
septation observed in this experiment are far lower, and, given the similar levels in nuc2+ and
nuc2-663, most likely reflect slippage of the hydroxyurea arrest. This leads us to conclude that
Nuc2p does not play a major role in preventing septation in interphase, once the M-G1 transi-
tion has been completed.
Contrary to traditional studies, where models are constructed from available data and are
then used for the experimental design of predictive simulations, our modeling approach is bidi-
rectional: in vivo experiments were performed to choose among refinement strategies during
model optimization, as well as the model was used to predict experimental outcomes (Fig 1).
The case of nuc2 regulation is an example of the former. Keeping all nuc2 SIN-related prior
knowledge in the multi-node CDK level model required the presence of a dual inhibitory con-
trol of the SIN in interphase by both Byr4p-Cdc16p and Nuc2p. In vivo experiments were per-
formed to identify the events that can be described by the model and, consequently, guide its
refinement strategy. The in vivo data argue against a post START role for Nuc2p, in addition to
that ascribed to it at the M-G1 transition. Therefore, nuc2 was removed from the final, model
presented here, which does not describe the M-G1 transition in its current form.
Fin1p over-expression may contribute to inactivation of the GAP for
Spg1p at mitotic onset
Fission yeast has a single orthologue of the conserved never-in-mitosis (nimA) kinase, called
fin1 [86]. Fin1p is not essential, but is important for spindle formation and regulates the affin-
ity of Plo1p to the SPB [87]. Fin1 mutant cells are delayed in the G2-M transition and Fin1p is
in part regulated by Sid2p [88]. This link between fin1 and the SIN prompted us to include fin1
in the SIN regulatory circuit.
In the PKN of the model there were no negative regulators targeting GAP components dur-
ing early mitosis, which resulted in suboptimal outcomes during the simulations of early mito-
sis; i.e. the simulation would produce a steady state where the GAP remained active in early
mitosis. Since removal of the SIN GAP from the SPB is an early step in the activation of the
SIN after entry into mitosis [12,74,89], we modeled whether GAP components could be regu-
lated by fin1. Since Cdc16p contains several sites matching the established consensus for mam-
malian Nek2 (one of the orthologues of nimA), the effect of increased expression of fin1 on
Cdc16p localization was investigated by in vivo experiments. Expression of fin1 from the
medium strength nmt-41 promoter [80] resulted in partial displacement of Cdc16p-GFP from
SPBs in interphase cells (Fig 5A). Quantification of the SPB-associated signal of Cdc16p-GFP
in interphase cells revealed that it was significantly decreased upon expression of fin1 (Fig 5B).
This was not due to a significant alteration of the steady state level of Cdc16p (Fig 5C).
Interestingly, the decreased level of Cdc16p-GFP at the SPB resulted in 24% of REP41-fin1
cells forming one or more septa in interphase, compared to <1% in the empty-vector control.
This is consistent with previous studies [73,85], which demonstrated that inactivation of cdc16-
116 in interphase cells promotes septation in interphasic cells. Therefore, the reduced level of
Cdc16p-GFP at the SPB may decrease the extent to which the SIN is inhibited by lowering the
amount of GAP available to inhibit Spg1p signaling.
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
11 / 22
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
12 / 22
Previous studies have shown that in the absence of GAP function, Fin1p acts as an inhibitor
of the SIN [85]. This study suggests that increased levels of Fin1p result in the reduction of
Cdc16p levels at the SPB, and therefore potentially to the activation of the SIN at the entry into
mitosis. This may point to a dual role of Fin1p in SIN regulation, which will be addressed in
future studies. Fin1p is implicated both in mitotic commitment, and in SIN regulation [85,88].
Expression of fin1 promotes recruitment of Plo1p to the SPB in interphase cells [87], and
Plo1p is involved in the displacement of Byr4p from the SPB in anaphase [46]. Future studies
will examine the mechanism by which Fin1p contributes to the decrease in Cdc16p at the SPB.
An unexpected prediction: cells overexpressing Cdc7p will septate in the
absence of Cdc11p or Sid4p
The final, optimized model describes the existing knowledge of the SIN, in wild type and known
mutants. One of the main goals of developing this Boolean model was to use it predictively by
performing in silico perturbations of interesting and/or experimentally challenging mutants.
The regulatory relationships described in this model predict that increased expression of Cdc7p
should produce septation in the absence of Cdc11p and Sid4p (Fig 4A). Previous studies have
shown that Spg1p overexpression will induce septation and permit colony formation in a cdc11
mutant [13], but not a sid4 mutant [91]. Moreover, increased expression of Cdc7p will permit
cdc11 mutants to form colonies [79]. In contrast to the situation with Spg1p overexpression,
induction of Cdc7p expression from the very strong nmt1 promoter in sid4-SA1 at 36°C did not
permit colony formation, but septa were formed in the cells (Fig 4B). To test whether increased
expression of Cdc7p would permit growth of a sid4 mutant, cdc7 was expressed from the ADH1
promoter, integrated at leu1. The leu1::pADH1-cdc7 strain has a very high septation index at
19°C (>90%) and is barely capable of colony formation at 25°C and above (Fig 4D), with cells
dying multiseptated at higher temperatures (Fig 4C). The strain sid4-SA1 leu1::pADH1-cdc7 was
capable of colony formation at 27°C and 29°C (Fig 4D), where neither parental strain could do
so. Previous studies have shown that increased expression of cdc7 increases the level of kinase
activity in immunoprecipitates of Cdc7p [79]. This shows that septation can occur if the function
of the scaffold proteins is compromised, provided the expression of Cdc7p is sufficiently elevated.
This raises the intriguing possibility that SIN signaling in this case originates in the cytoplasm,
bypassing the need for assembly on a SPB-associated scaffold. The nature of the SIN protein sig-
naling complexes present in these cells will be the subject of future studies.
Discussion
In this paper we use qualitative Boolean modeling to represent and explore the regulatory rela-
tionships of genes participating in the Septation Initiation Network of fission yeast. Qualitative
Fig 5. Fin1p over-expression results in Cdc16p disassociation from the SPB. (A) Cells expressing the
labeled tubulin marker leu1::m-Cherry-atb2 and cdc16-GFP were induced to express fin1 from the medium
strength nmt41 promoter [80]. Cells transformed with empty vector served as control. Cells were grown in
medium without thiamine for 27h at 25°C. Cells were imaged and the intensity of SPB associated cdc16-GFP
signal was analyzed as described in [12]. The panel shows m-Cherry-atb2 leu1::cdc16-GFP(ura4+) cells
bearing REP41 or REP41-fin1. The scale bar is 10 μm. (B) The SPB associated signal was determined in
interphase cells in each strain. Since REP41-fin1 eventually leads to a mitotic arrest [87] interphase cells
were identified by the presence of an interphasic microtubule array. The box shows 25%-75% range for the
population, the line indicates the median. The bars indicate 10% and 90% range for the population, and dots
indicate more extreme individual values. The y-axis shows fluorescence intensity on an arbitrary scale. (C)
Cells bearing the leu1::cdc16-HA allele were induced to express fin1 (ON) by growing them in defined
minimal medium [81] in the presence (OFF) or absence (ON) of 2mM thiamine. Protein extracts were
prepared 27h after induction and analyzed by western blotting using monoclonal antibody 12CA5. The anti-α-
tubulin monoclonal antibody TAT-1 [90] was used as a control.
doi:10.1371/journal.pone.0134214.g005
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
13 / 22
modeling is a powerful method for systems with restricted kinetic information and it is compu-
tationally efficient, allowing for thousands of in silico experiments in a short time, even in net-
works with hundreds of nodes. Moreover, it can be used predictively, to test combination of
mutations that would otherwise be time consuming, expensive and/or experimentally challeng-
ing to undertake. The value of such models increases significantly when the model is coupled
with in vivo experiments. Such experiments can be used to evaluate the regulatory rules, help
the optimization procedure and test the predictions of the model (Fig 1).
We report the construction of an extended, Boolean model of the SIN network that uses
CDK levels as control nodes to simulate SIN related events in interphase, early mitosis and late
mitosis. The prior knowledge network was manually curated, providing a trustworthy initial
framework that could then be further optimized (Fig 1). Information reported in literature
(and used in network databases) can be conflicting, outdated, incomplete or based on in vitro
knowledge only. Therefore, expert biocuration provides a significant advantage in order to fil-
ter the available information and construct a comprehensive network.
We optimized the model using in silico experiments with well-established outcomes based
on in vivo data, in order to recapitulate the SIN state in different stages of the cell cycle (Fig 1).
A challenging aspect of qualitative modeling, and especially of asynchronous update, is to
interpret the resulting steady states of the simulations. This is because the simulation might
result in a number of steady states that are theoretically possible but never reached in vivo. Our
approach was to use CDK levels as an initial condition for the simulation, indicating the stage
of the cell cycle that the simulation corresponds, to reduce unrealistic simulation outcomes.
We further restricted the simulation space by taking as a fact that the scaffold has the potential
to be constructed at all times by setting the SIN-SPB linker protein Ppc89p to 1.
The optimization process under the controlled environment of CDK switches provided
important insights into SIN regulation during the cell cycle. In the case of the fin1, the incorrect
simulation results that were obtained in early mitosis helped us locate a potential missing link
in the PKN. Increased expression of fin1 removes Cdc16p from the SPB. At present we do not
know whether this is by direct phosphorylation of Cdc16p or an indirect effect; this will be the
subject of future analysis. However, the important point in this context is that the modeling
revealed the requirement for an additional control point to turn off the GAP in early mitosis.
The optimization strategy was also useful in evaluating the limitations of our model. An exam-
ple of this is the role of Nuc2p in SIN regulation. In the PKN there were several inhibitory links
from Nuc2p to SIN kinases, indicating the events in SIN resetting, after septation [84]. The use
of CDK switches restricts the cell cycle events that can be modeled, and our model does not
presently incorporate resetting of the SIN at the M-G1 transition. Our modeling predicted that
if Nuc2p continued to activate the GAP in interphase, extending the role proposed for it at the
M-G1 transition [84], then its inactivation in post-START cells could result in septum forma-
tion; in vivo analysis showed this was not the case. Thus, the modeling was useful in this case to
define the possible limits of the extent of the time-window in which Nuc2p is active towards
the SIN.
The great value of creating an optimized qualitative model is that it can then be used predic-
tively to perform difficult or iconoclastic experiments in silico. We focused on testing whether
an over-expression of SIN kinases would rescue SIN scaffold mutants. The model’s prediction
was that over-expression of Cdc7p in a cdc11 or sid4 knock-out will still septate, a prediction
that was experimentally validated. The model can be used in the future for any combination of
gene mutants, and hopefully provide interesting hypotheses that can be tested experimentally.
Future studies will aim to model the M-G1 transition, and to incorporate spatial components
into the model (protein localization to one or both SPB, cytoplasm or division site). This will
be facilitated by the incorporation of the cell cycle Boolean module of fission yeast, by Davidich
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
14 / 22
& Bornholdt [30]. We will also incorporate multivariate nodes to simulate the effect of changes
in the post-translational modifications of SIN proteins during the cell cycle [41,92,93]. This
should allow modeling of the role of the asymmetry of SPBs with regard to SIN protein associa-
tion, building upon the analysis performed by Bajpai et al. [41]. Future versions of the model
will attempt to incorporate Etd1p. Though its effects upon SIN signaling are clear, the pub-
lished analyses do not provide a sufficiently clear, direct link to SIN components to permit its
unequivocal incorporation into the model presented here.
Our extended, Boolean model of the SIN can be used by the scientific community for testing
various hypotheses in silico, including multiple gene perturbations that can be experimentally
challenging. The model can be reduced to a minimum number of nodes and still capture the
system steady states (see S3 File). Though a reductive approach can be a useful aid in under-
standing the information flow in the system, the greater complexity of the extended model sys-
tem increases the predictive value of the model, as we can use the model nodes for testing the
desired experimental scenarios.
Finally, it is worth noting that qualitative models such as the one presented here are over-
simplifications of the actual regulatory processes; in our case of the regulation of the SIN. With
advances in live monitoring of cell division and development of new fluorescent probes, we
should be able to generate more accurate quantitative models for such a system. Our approach
is nevertheless an important step towards a more comprehensive model that recapitulates
known biology of the SIN and can be used as a hypothesis generator for complex experimental
design.
Materials and Methods
Literature database construction
For the construction of the PKN, several online resources were curated to retrieve SIN relevant
information, such as Pathguide, Pubmed, iHOP, iRefWeb, Scholar Google, PriME and Uni-
ProtKB/Swiss-Prot. The collected information was stored in a structure formed of pairwise
interactions and regulations that includes information about participating components, the
origin of publications (PMID) where the interaction was mentioned and a confidence level as
an evidence tag from the biocurator. In detail, the database contains the following columns:
1. Node 1: The name of the first element of the interaction, the one that acts as activator or
inhibitor
2. Action: A symbol characterizing the type of interaction as activation (->) or inhibition (-|)
3. Node 2: The name of the second element of the interaction, the one that gets activated or
inhibited
4. Node 1 type: The type of node 1. Can be protein, complex or miRNA
5. Node 2 type: The type of node 2. Can be protein or miRNA
6. UniProt ID 1: Reference of node 1
7. UniProt ID 2: Reference of node 2
8. PMID: Literature reference of the interaction
9. Class: A letter characterizing the confidence level of the interaction. It can be one of the
following:
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
15 / 22
Sure (S), when the interaction is confirmed or known in textbook, and/or already in the Uni-
Prot general annotation lines. Sure interactions are generally associated with many PMIDs.
Unsure (U), when the interaction is shown once and/or not confirmed by others, or when the
authors are not confident about the results.
Inferred (I), when there are no results for the network in question, but the interaction was
found in different cell types and/or organisms. It may also refer to cases where the informa-
tion is inferred to all protein isoforms of a gene without confirmed results. Inferred interac-
tions might be associated with more than one PMIDs.
Contradictory (C), when the interaction is based on contradictory results.
10. Evidence tag: Short extract from the publication where the interaction is mentioned.
GenYsis Boolean modeling toolbox
All Boolean simulations of the SIN model, including the identification of wild type attractors
and in silico perturbation experiments, we performed using the genYsis Boolean modeling tool-
box. GenYsis uses reduced ordered binary decision diagrams (ROBDDs) in order to efficiently
compute attractors and steady states of large networks. ROBDDs are directed acyclic graphs
that can represent Boolean functions efficiently, and are computationally suitable for complex
Boolean operations. To map gene regulatory networks on ROBDDs the network has to be
transformed into Boolean functions that represent the dynamics of the model. All the opera-
tions that can be performed on Boolean functions can also be performed on their correspond-
ing ROBDD representations [69]. The simulation modes available with genYsis include
synchronous and asynchronous updating. In both cases, the user has the possibility of perform-
ing in silico perturbations by fixing the activation state of one or multiple components during
simulation. The perturbation possibilities comprise (a) knock-out experiments, were the
selected components are set to be inactive during the whole simulation, (b) over-expression
experiments, were the selected components are set to be active during the whole simulation,
and (c) initial state experiments, where the selected components have a fixed activation state at
the beginning of the simulation that is thereafter allowed to change according to the regulatory
rules. The software binaries of genYsis are available for Linux (64 bits gcc version 4.4.5, Debian
4.4.5–8) and Mac OS X (64 bits gcc version 4.2.1, Mac OS X 10.8.5) at http://www.vital-it.ch/
software/genYsis.
Fission yeast techniques
(A) Media.
Growth and manipulation of S. pombe was performed according to standard
protocols [81]. Defined medium was EMM2 with supplements at 100 mg/l as required,
and complete medium was YE [81]. Cell number was determined using a hemocytometer.
For the induction of nmt1 regulated genes, cells were grown to exponential phase (approx.
2-3x 106 ml-1) in EMM2 containing 2 μM thiamine with additional supplements as required.
Cells were washed twice in medium without thiamine, and then grown for the time indicated
in the Fig legends.
(B) Molecular and Genetic analyses.
Strains were constructed by standard genetic meth-
ods. Vectors [80,94] and cdc7 plasmids [79] have been described previously.
(C) Imaging and image analysis.
Living cells were imaged using a U-Plan-S-Apo 60× N.
A. 1.42 objective lens mounted on an Olympus IX-81 spinning disc confocal microscope. The
temperature was maintained using a custom-built heating system. Fixed cells were photo-
graphed on a Zeiss Axiophot microscope using a Zeiss 100x NA 1.4 PLAN-apochromat lens.
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
16 / 22
Images were captured on a Nikon Coolpix camera. Level adjustment and cropping were per-
formed using Adobe Photoshop CS6.
DAPI and Calcofluor staining was performed on cells that had been harvested by centrifu-
gation, washed, and fixed with cold 70% (v/v) ethanol, as described previously [95]. Micros-
copy analysis of living cells was performed as described in [12], using the RodcellJ imageJ
plugin [96]. Data were plotted using GraphPad Prism v6. In the whisker plots the box shows
25%-75% range for the population, the line indicates the median. The bars indicate 10% and
90% range for the population, and dots indicate more extreme individual values.
Supporting Information
S1 File. The SIN model in genYsis format. The final model in genYsis format can be used for
any combination of in silico experiments using the genYsis software.
(ZIP)
S2 File. The SIN model in SMBL qual format. The final model in SBML qual format can be
used to perform additional analyses in most qualitative modeling platforms.
(ZIP)
S3 File. Model reduction analysis. A reduction analysis performed using GINsim [97],
highlighting the information flow that is necessary for the maintenance of the system’s steady
states.
(PDF)
S1 Fig. The model optimization scoring set. The experiments used to score the model candi-
dates during the optimization phase are represented in the y axis and the proteins used for
scoring in the x axis. The table uses the same color coding as the article figures: blue for Bool-
ean state 1, white for Boolean state 0 and light blue for oscillation or, in this case, two alterna-
tive steady states with different activation states of the given protein.
(TIF)
S2 Fig. Detailed in silico results of final model simulations. A detailed heatmap showing the
activation state of all nodes of the final model for all experiments presented in this paper.
(TIF)
S1 Table. Prior Knowledge Network interaction table. A complete list of the interactions
included in the Prior Knowledge Network of the SIN.
(PDF)
S2 Table. Final model interaction list. The list of interactions comprising the final, optimized
model, together on comments justifying their alteration / addition.
(PDF)
Acknowledgments
We thank Iain Hagan (Paterson institute for Cancer Research, Manchester, UK) for the fin1
expression plasmids and Keith Gull (Oxford, UK), for TAT-1. A special thanks to Julien Dorier
(SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland) for the constructive discussions
all along this project.
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
17 / 22
Author Contributions
Conceived and designed the experiments: IX VS. Performed the experiments: AC PW AN VS
PC AK EC. Analyzed the data: AC PW PC. Wrote the paper: AC VS PW IX.
References
1.
Bardin AJ, Amon A (2001) Men and sin: what's the difference? Nat Rev Mol Cell Biol 2: 815–826.
PMID: 11715048
2.
Seshan A, Amon A (2004) Linked for life: temporal and spatial coordination of late mitotic events. Curr
Opin Cell Biol 16: 41–48. PMID: 15037303
3.
Chang L, Morrell JL, Feoktistova A, Gould KL (2001) Study of cyclin proteolysis in anaphase-promoting
complex (APC) mutant cells reveals the requirement for APC function in the final steps of the fission
yeast septation initiation network. Mol Cell Biol 21: 6681–6694. PMID: 11533255
4.
Guertin DA, Chang L, Irshad F, Gould KL, McCollum D (2000) The role of the sid1p kinase and cdc14p
in regulating the onset of cytokinesis in fission yeast. EMBO J 19: 1803–1815. PMID: 10775265
5.
Simanis V (2015) Pombe's thirteen—control of fission yeast cell division by the septation initiation net-
work. J Cell Sci 128: 1465–1474. doi: 10.1242/jcs.094821 PMID: 25690009
6.
Mitchison JM, Nurse P (1985) Growth in cell length in the fission yeast Schizosaccharomyces pombe. J
Cell Sci 75: 357–376. PMID: 4044680
7.
Minet M, Nurse P, Thuriaux P, Mitchison JM (1979) Uncontrolled septation in a cell division cycle
mutant of the fission yeast Schizosaccharomyces pombe. J Bacteriol 137: 440–446. PMID: 762020
8.
Tomlin GC, Morrell JL, Gould KL (2002) The spindle pole body protein Cdc11p links Sid4p to the fission
yeast septation initiation network. Mol Biol Cell 13: 1203–1214. PMID: 11950932
9.
Krapp A, Schmidt S, Cano E, Simanis V (2001) S. pombe cdc11p, together with sid4p, provides an
anchor for septation initiation network proteins on the spindle pole body. Curr Biol 11: 1559–1568.
PMID: 11676915
10.
Rosenberg JA, Tomlin GC, McDonald WH, Snydsman BE, Muller EG, et al. (2006) Ppc89 links multiple
proteins, including the septation initiation network, to the core of the fission yeast spindle-pole body.
Mol Biol Cell 17: 3793–3805. PMID: 16775007
11.
Chang L, Gould KL (2000) Sid4p is required to localize components of the septation initiation pathway
to the spindle pole body in fission yeast. Proc Natl Acad Sci U S A 97: 5249–5254. PMID: 10805785
12.
Wachowicz P, Chasapi A, Krapp A, Cano Del Rosario E, Schmitter D, et al. (2015) Analysis of S.
pombe SIN protein association to the SPB reveals two genetically separable states of the SIN. J Cell
Sci 128: 741–754. doi: 10.1242/jcs.160150 PMID: 25501816
13.
Schmidt S, Sohrmann M, Hofmann K, Woollard A, Simanis V (1997) The Spg1p GTPase is an essen-
tial, dosage-dependent inducer of septum formation in Schizosaccharomyces pombe. Genes Dev 11:
1519–1534. PMID: 9203579
14.
Mehta S, Gould KL (2006) Identification of functional domains within the septation initiation network
kinase, Cdc7. J Biol Chem 281: 9935–9941. PMID: 16469735
15.
Guertin DA, McCollum D (2001) Interaction between the noncatalytic region of Sid1p kinase and
Cdc14p is required for full catalytic activity and localization of Sid1p. J Biol Chem 276: 28185–28189.
PMID: 11384993
16.
Salimova E, Sohrmann M, Fournier N, Simanis V (2000) The S. pombe orthologue of the S. cerevisiae
mob1 gene is essential and functions in signalling the onset of septum formation. J Cell Sci 113 (Pt
10): 1695–1704.
17.
Hou MC, Salek J, McCollum D (2000) Mob1p interacts with the Sid2p kinase and is required for cytoki-
nesis in fission yeast. Curr Biol 10: 619–622. PMID: 10837231
18.
Hou MC, Guertin DA, McCollum D (2004) Initiation of cytokinesis is controlled through multiple modes
of regulation of the Sid2p-Mob1p kinase complex. Mol Cell Biol 24: 3262–3276. PMID: 15060149
19.
Johnson AE, McCollum D, Gould KL (2012) Polar opposites: Fine-tuning cytokinesis through SIN
asymmetry. Cytoskeleton (Hoboken) 69: 686–699. doi: 10.1002/cm.21044 PMID: 22786806
20.
Simanis V (2003) The mitotic exit and septation initiation networks. J Cell Sci 116: 4261–4262. PMID:
14514881
21.
Furge KA, Wong K, Armstrong J, Balasubramanian M, Albright CF (1998) Byr4 and Cdc16 form a two-
component GTPase-activating protein for the Spg1 GTPase that controls septation in fission yeast.
Curr Biol 8: 947–954. PMID: 9742395
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
18 / 22
22.
Furge KA, Cheng QC, Jwa M, Shin S, Song K, et al. (1999) Regions of Byr4, a regulator of septation in
fission yeast, that bind Spg1 or Cdc16 and form a two-component GTPase-activating protein with
Cdc16. J Biol Chem 274: 11339–11343. PMID: 10196225
23.
Alcaide-Gavilan M, Lahoz A, Daga RR, Jimenez J (2014) Feedback regulation of SIN by Etd1 and
Rho1 in fission yeast. Genetics 196: 455–470. doi: 10.1534/genetics.113.155218 PMID: 24336750
24.
Daga RR, Lahoz A, Munoz MJ, Moreno S, Jimenez J (2005) Etd1p is a novel protein that links the SIN
cascade with cytokinesis. EMBO J 24: 2436–2446. PMID: 15933715
25.
Garcia-Cortes JC, McCollum D (2009) Proper timing of cytokinesis is regulated by Schizosaccharo-
myces pombe Etd1. J Cell Biol 186: 739–753. doi: 10.1083/jcb.200902116 PMID: 19736319
26.
Lahoz A, Alcaide-Gavilan M, Daga RR, Jimenez J (2010) Antagonistic roles of PP2A-Pab1 and Etd1 in
the control of cytokinesis in fission yeast. Genetics 186: 1261–1270. doi: 10.1534/genetics.110.
121368 PMID: 20876564
27.
Krapp A, Cano E, Simanis V (2003) Mitotic hyperphosphorylation of the fission yeast SIN scaffold pro-
tein cdc11p is regulated by the protein kinase cdc7p. Curr Biol 13: 168–172. PMID: 12546793
28.
Tanaka K, Petersen J, MacIver F, Mulvihill DP, Glover DM, et al. (2001) The role of Plo1 kinase in
mitotic commitment and septation in Schizosaccharomyces pombe. EMBO J 20: 1259–1270. PMID:
11250892
29.
Goyal A, Takaine M, Simanis V, Nakano K (2011) Dividing the spoils of growth and the cell cycle: The
fission yeast as a model for the study of cytokinesis. Cytoskeleton (Hoboken) 68: 69–88.
30.
Davidich MI, Bornholdt S (2008) Boolean network model predicts cell cycle sequence of fission yeast.
PLoS One 3: e1672. doi: 10.1371/journal.pone.0001672 PMID: 18301750
31.
Friedman N, Linial M, Nachman I, Pe'er D (2000) Using Bayesian networks to analyze expression data.
J Comput Biol 7: 601–620. PMID: 11108481
32.
Tyson JJ, Csikasz-Nagy A, Novak B (2002) The dynamics of cell cycle regulation. Bioessays 24:
1095–1109. PMID: 12447975
33.
Kahlem P, DiCara A, Durot M, Hancock JM, Klipp E, et al. (2011) Strengths and Weaknesses of
Selected Modeling Methods Used in Systems Biology. In: Y N.S., editor. Systems and Computational
Biology InTech.
34.
Wang RS, Saadatpour A, Albert R (2012) Boolean modeling in systems biology: an overview of meth-
odology and applications. Phys Biol 9: 055001. doi: 10.1088/1478-3975/9/5/055001 PMID: 23011283
35.
Chen KC, Calzone L, Csikasz-Nagy A, Cross FR, Novak B, et al. (2004) Integrative analysis of cell
cycle control in budding yeast. Mol Biol Cell 15: 3841–3862. PMID: 15169868
36.
Chen KC, Csikasz-Nagy A, Gyorffy B, Val J, Novak B, et al. (2000) Kinetic analysis of a molecular
model of the budding yeast cell cycle. Mol Biol Cell 11: 369–391. PMID: 10637314
37.
Faure A, Naldi A, Lopez F, Chaouiya C, Ciliberto A, et al. (2009) Modular logical modelling of the bud-
ding yeast cell cycle. Mol Biosyst 5: 1787–1796. doi: 10.1039/B910101m PMID: 19763337
38.
Ingalls BP, Duncker BP, Kim DR, McConkey BJ (2007) Systems level modeling of the cell cycle using
budding yeast. Cancer Inform 3: 357–370. PMID: 19455254
39.
Irons DJ (2009) Logical analysis of the budding yeast cell cycle. J Theor Biol 257: 543–559. doi: 10.
1016/j.jtbi.2008.12.028 PMID: 19185585
40.
Li F, Long T, Lu Y, Ouyang Q, Tang C (2004) The yeast cell-cycle network is robustly designed. Proc
Natl Acad Sci U S A 101: 4781–4786. PMID: 15037758
41.
Bajpai A, Feoktistova A, Chen JS, McCollum D, Sato M, et al. (2013) Dynamics of SIN asymmetry
establishment. PLoS Comput Biol 9: e1003147. doi: 10.1371/journal.pcbi.1003147 PMID: 23874188
42.
Csikasz-Nagy A, Kapuy O, Gyorffy B, Tyson JJ, Novak B (2007) Modeling the septation initiation net-
work (SIN) in fission yeast cells. Curr Genet 51: 245–255. PMID: 17340144
43.
Bateman A (2010) Curators of the world unite: the International Society of Biocuration. Bioinformatics
26: 991. doi: 10.1093/bioinformatics/btq101 PMID: 20305270
44.
Poux S, Magrane M, Arighi CN, Bridge A, O'Donovan C, et al. (2014) Expert curation in UniProtKB: a
case study on dealing with conflicting and erroneous data. Database (Oxford) 2014: bau016.
45.
Krapp A, Collin P, Cano Del Rosario E, Simanis V (2008) Homoeostasis between the GTPase Spg1p
and its GAP in the regulation of cytokinesis in S. pombe. J Cell Sci 121: 601–608. doi: 10.1242/jcs.
022772 PMID: 18252797
46.
Rachfall N, Johnson AE, Mehta S, Chen JS, Gould KL (2014) Cdk1 Promotes Cytokinesis in Fission
Yeast through Activation of the Septation Initiation Network. Mol Biol Cell.
47.
Akutsu T, Miyano S, Kuhara S (1999) Identification of genetic networks from a small number of gene
expression patterns under the Boolean network model. Pac Symp Biocomput: 17–28. PMID: 10380182
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
19 / 22
48.
Garg A, Mohanram K, De Micheli G, Xenarios I (2012) Implicit methods for qualitative modeling of gene
regulatory networks. Methods Mol Biol 786: 397–443. doi: 10.1007/978-1-61779-292-2_22 PMID:
21938638
49.
Garg A, Mohanram K, Di Cara A, De Micheli G, Xenarios I (2009) Modeling stochasticity and robust-
ness in gene regulatory networks. Bioinformatics 25: i101–109. doi: 10.1093/bioinformatics/btp214
PMID: 19477975
50.
Kauffman SA (1993) The origins of order: self-organization and selection in evolution. New York;
Oxford: Oxford University Press. xviii, 709 p p.
51.
Kochi N, Matache MT (2012) Mean-field Boolean network model of a signal transduction network. Bio-
systems 108: 14–27. doi: 10.1016/j.biosystems.2011.12.001 PMID: 22212351
52.
Albert R, Othmer HG (2003) The topology of the regulatory interactions predicts the expression pattern
of the segment polarity genes in Drosophila melanogaster. J Theor Biol 223: 1–18. PMID: 12782112
53.
Azpeitia E, Benitez M, Padilla-Longoria P, Espinosa-Soto C, Alvarez-Buylla ER (2011) Dynamic net-
work-based epistasis analysis: boolean examples. Front Plant Sci 2: 92. doi: 10.3389/fpls.2011.00092
PMID: 22645556
54.
Giacomantonio CE, Goodhill GJ (2010) A Boolean model of the gene regulatory network underlying
Mammalian cortical area development. PLoS Comput Biol 6.
55.
Li S, Assmann SM, Albert R (2006) Predicting essential components of signal transduction networks: a
dynamic model of guard cell abscisic acid signaling. PLoS Biol 4: e312. PMID: 16968132
56.
Morris MK, Saez-Rodriguez J, Sorger PK, Lauffenburger DA (2010) Logic-based models for the analy-
sis of cell signaling networks. Biochemistry 49: 3216–3224. doi: 10.1021/bi902202q PMID: 20225868
57.
Saez-Rodriguez J, Simeoni L, Lindquist JA, Hemenway R, Bommhardt U, et al. (2007) A logical model
provides insights into T cell receptor signaling. PLoS Comput Biol 3: e163. PMID: 17722974
58.
Samaga R, Saez-Rodriguez J, Alexopoulos LG, Sorger PK, Klamt S (2009) The logic of EGFR/ErbB
signaling: theoretical properties and analysis of high-throughput data. PLoS Comput Biol 5: e1000438.
doi: 10.1371/journal.pcbi.1000438 PMID: 19662154
59.
Sanchez L, van Helden J, Thieffry D (1997) Establishement of the dorso-ventral pattern during embry-
onic development of drosophila melanogasater: a logical analysis. J Theor Biol 189: 377–389. PMID:
9446747
60.
Schlatter R, Philippi N, Wangorsch G, Pick R, Sawodny O, et al. (2012) Integration of Boolean models
exemplified on hepatocyte signal transduction. Brief Bioinform 13: 365–376. doi: 10.1093/bib/bbr065
PMID: 22016404
61.
Schlatter R, Schmich K, Avalos Vizcarra I, Scheurich P, Sauter T, et al. (2009) ON/OFF and beyond—a
boolean model of apoptosis. PLoS Comput Biol 5: e1000595. doi: 10.1371/journal.pcbi.1000595
PMID: 20011108
62.
Veliz-Cuba A, Stigler B (2011) Boolean models can explain bistability in the lac operon. J Comput Biol
18: 783–794. doi: 10.1089/cmb.2011.0031 PMID: 21563979
63.
Naldi A, Thieffry D, Chaouiya C (2007) Decision Diagrams for the Representation and Analysis of Logi-
cal Models of Genetic Networks. In: Calder M, Gilmore S, editors. Computational Methods in Systems
Biology: Springer Berlin Heidelberg. pp. 233–247.
64.
Saadatpour A, Wang RS, Liao A, Liu X, Loughran TP, et al. (2011) Dynamical and structural analysis of
a T cell survival network identifies novel candidate therapeutic targets for large granular lymphocyte
leukemia. PLoS Comput Biol 7: e1002267. doi: 10.1371/journal.pcbi.1002267 PMID: 22102804
65.
Chaves M, Sontag ED, Albert R (2006) Methods of robustness analysis for Boolean models of gene
control networks. Syst Biol (Stevenage) 153: 154–167.
66.
Chaves M, Albert R, Sontag ED (2005) Robustness and fragility of Boolean models for genetic regula-
tory networks. J Theor Biol 235: 431–449. PMID: 15882705
67.
Faure A, Naldi A, Chaouiya C, Thieffry D (2006) Dynamical analysis of a generic Boolean model for the
control of the mammalian cell cycle. Bioinformatics 22: e124–131. PMID: 16873462
68.
Helikar T, Rogers JA (2009) ChemChains: a platform for simulation and analysis of biochemical net-
works aimed to laboratory scientists. BMC Syst Biol 3: 58. doi: 10.1186/1752-0509-3-58 PMID:
19500393
69.
Garg A, Di Cara A, Xenarios I, Mendoza L, De Micheli G (2008) Synchronous versus asynchronous
modeling of gene regulatory networks. Bioinformatics 24: 1917–1925. doi: 10.1093/bioinformatics/
btn336 PMID: 18614585
70.
Dischinger S, Krapp A, Xie L, Paulson JR, Simanis V (2008) Chemical genetic analysis of the regulatory
role of Cdc2p in the S. pombe septation initiation network. J Cell Sci 121: 843–853. doi: 10.1242/jcs.
021584 PMID: 18303049
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
20 / 22
71.
He X, Patterson TE, Sazer S (1997) The Schizosaccharomyces pombe spindle checkpoint protein
mad2p blocks anaphase and genetically interacts with the anaphase-promoting complex. Proc Natl
Acad Sci U S A 94: 7965–7970. PMID: 9223296
72.
Yamano H, Gannon J, Hunt T (1996) The role of proteolysis in cell cycle progression in Schizosacchar-
omyces pombe. EMBO J 15: 5268–5279. PMID: 8895572
73.
Cerutti L, Simanis V (1999) Asymmetry of the spindle pole bodies and spg1p GAP segregation during
mitosis in fission yeast. J Cell Sci 112 (Pt 14): 2313–2321.
74.
Baum B, Wuarin J, Nurse P (1997) Control of S-phase periodic transcription in the fission yeast mitotic
cycle. EMBO J 16: 4676–4688. PMID: 9303312
75.
Coudreuse D, Nurse P (2010) Driving the cell cycle with a minimal CDK control network. Nature 468:
1074–1079. doi: 10.1038/nature09543 PMID: 21179163
76.
Grallert A, Patel A, Tallada VA, Chan KY, Bagley S, et al. (2013) Centrosomal MPF triggers the mitotic
and morphogenetic switches of fission yeast. Nat Cell Biol 15: 88–95. doi: 10.1038/ncb2633 PMID:
23222840
77.
Chaouiya C, Berenguier D, Keating SM, Naldi A, van Iersel MP, et al. (2013) SBML qualitative models:
a model representation format and infrastructure to foster interactions between qualitative modelling
formalisms and tools. BMC Syst Biol 7: 135. doi: 10.1186/1752-0509-7-135 PMID: 24321545
78.
Marks J, Fankhauser C, Simanis V (1992) Genetic interactions in the control of septation in Schizosac-
charomyces pombe. J Cell Sci 101 (Pt 4): 801–808.
79.
Fankhauser C, Simanis V (1994) The cdc7 protein kinase is a dosage dependent regulator of septum
formation in fission yeast. EMBO J 13: 3011–3019. PMID: 8039497
80.
Basi G, Schmid E, Maundrell K (1993) TATA box mutations in the Schizosaccharomyces pombe nmt1
promoter affect transcription efficiency but not the transcription start point or thiamine repressibility.
Gene 123: 131–136. PMID: 8422997
81.
Moreno S, Klar A, Nurse P (1991) Molecular genetic analysis of fission yeast Schizosaccharomyces
pombe. Methods Enzymol 194: 795–823. PMID: 2005825
82.
Hirano T, Hiraoka Y, Yanagida M (1988) A temperature-sensitive mutation of the Schizosaccharo-
myces pombe gene nuc2+ that encodes a nuclear scaffold-like protein blocks spindle elongation in
mitotic anaphase. J Cell Biol 106: 1171–1183. PMID: 3283148
83.
Kumada K, Su S, Yanagida M, Toda T (1995) Fission yeast TPR-family protein nuc2 is required for G1-
arrest upon nitrogen starvation and is an inhibitor of septum formation. J Cell Sci 108 (Pt 3): 895–905.
84.
Chew TG, Balasubramanian MK (2008) Nuc2p, a subunit of the anaphase-promoting complex, inhibits
septation initiation network following cytokinesis in fission yeast. PLoS Genet 4: e17. doi: 10.1371/
journal.pgen.0040017 PMID: 18225957
85.
Grallert A, Krapp A, Bagley S, Simanis V, Hagan IM (2004) Recruitment of NIMA kinase shows that
maturation of the S. pombe spindle-pole body occurs over consecutive cell cycles and reveals a role for
NIMA in modulating SIN activity. Genes Dev 18: 1007–1021. PMID: 15132994
86.
Krien MJ, Bugg SJ, Palatsides M, Asouline G, Morimyo M, et al. (1998) A NIMA homologue promotes
chromatin condensation in fission yeast. J Cell Sci 111 (Pt 7): 967–976.
87.
Grallert A, Hagan IM (2002) Schizosaccharomyces pombe NIMA-related kinase, Fin1, regulates spin-
dle formation and an affinity of Polo for the SPB. EMBO J 21: 3096–3107. PMID: 12065422
88.
Grallert A, Connolly Y, Smith DL, Simanis V, Hagan IM (2012) The S. pombe cytokinesis NDR kinase
Sid2 activates Fin1 NIMA kinase to control mitotic commitment through Pom1/Wee1. Nat Cell Biol 14:
738–745. doi: 10.1038/ncb2514 PMID: 22684255
89.
Li C, Furge KA, Cheng QC, Albright CF (2000) Byr4 localizes to spindle-pole bodies in a cell cycle-regu-
lated manner to control Cdc7 localization and septation in fission yeast. J Biol Chem 275: 14381–
14387. PMID: 10799520
90.
Woods A, Sherwin T, Sasse R, MacRae TH, Baines AJ, et al. (1989) Definition of individual compo-
nents within the cytoskeleton of Trypanosoma brucei by a library of monoclonal antibodies. J Cell Sci
93 (Pt 3): 491–500.
91.
Balasubramanian MK, McCollum D, Chang L, Wong KC, Naqvi NI, et al. (1998) Isolation and character-
ization of new fission yeast cytokinesis mutants. Genetics 149: 1265–1275. PMID: 9649519
92.
Feoktistova A, Morrell-Falvey J, Chen JS, Singh NS, Balasubramanian MK, et al. (2012) The fission
yeast septation initiation network (SIN) kinase, Sid2, is required for SIN asymmetry and regulates the
SIN scaffold, Cdc11. Mol Biol Cell 23: 1636–1645. doi: 10.1091/mbc.E11-09-0792 PMID: 22419817
93.
Singh NS, Shao N, McLean JR, Sevugan M, Ren L, et al. (2011) SIN-inhibitory phosphatase complex
promotes Cdc11p dephosphorylation and propagates SIN asymmetry in fission yeast. Curr Biol 21:
1968–1978. doi: 10.1016/j.cub.2011.10.051 PMID: 22119525
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
21 / 22
94.
Fennessy D, Grallert A, Krapp A, Cokoja A, Bridge AJ, et al. (2014) Extending the Schizosaccharo-
myces pombe molecular genetic toolbox. PLoS One 9: e97683. doi: 10.1371/journal.pone.0097683
PMID: 24848109
95.
Balasubramanian MK, McCollum D, Gould KL (1997) Cytokinesis in fission yeast Schizosaccharo-
myces pombe. Methods Enzymol 283: 494–506. PMID: 9251043
96.
Schmitter D, Wachowicz P, Sage D, Chasapi A, Xenarios I, et al. (2013) A 2D/3D image analysis sys-
tem to track fluorescently labeled structures in rod-shaped cells: application to measure spindle pole
asymmetry during mitosis. Cell Div 8: 6. doi: 10.1186/1747-1028-8-6 PMID: 23622681
97.
Chaouiya C, Naldi A, Thieffry D (2012) Logical modelling of gene regulatory networks with GINsim.
Methods Mol Biol 804: 463–479. doi: 10.1007/978-1-61779-361-5_23 PMID: 22144167
Boolean Model of the S. pombe SIN
PLOS ONE | DOI:10.1371/journal.pone.0134214
August 5, 2015
22 / 22
|
26244885
|
dma1 = ( sid4 AND ( ( ( CK1 ) ) ) )
plo1 = ( ( ( cdk-H ) AND NOT ( sid4 AND ( ( ( dma1 ) ) ) ) ) AND NOT ( cdk-L ) ) OR ( ( sid4 ) AND NOT ( sid4 AND ( ( ( dma1 ) ) ) ) )
pom1 = ( pak1 )
sid2 = ( cdc11 AND ( ( ( cdc7 AND SIP ) ) ) ) OR ( cdc7 )
Rga4 = NOT ( ( pom1 ) )
fin1 = ( cdk-H )
SIP = ( ( cdk-H ) AND NOT ( cdk-0 AND ( ( ( ppc89 ) ) ) ) )
byr4-cdc16 = ( byr4 AND ( ( ( cdc16 ) ) ) )
byr4 = ( ( ( cdk-L ) AND NOT ( cdk-H AND ( ( ( plo1 ) ) ) ) ) AND NOT ( cdc42 ) )
cdc11 = ( cdc16 ) OR ( flp1 AND ( ( ( sid4 ) ) ) ) OR ( sid4 ) OR ( cdc7 )
cdc7 = ( spg1 AND ( ( ( cdc11 ) ) ) )
sid4 = ( ppc89 )
flp1 = ( ( sid2-mob1 ) AND NOT ( cdk-H ) ) OR ( ( cdk-0 ) AND NOT ( cdk-H ) )
scd1 = ( ras1 )
spg1 = ( ( cdc11 ) AND NOT ( byr4-cdc16 ) ) OR ( ( etd1 ) AND NOT ( byr4-cdc16 ) )
gef1 = ( orb6 )
cdc16 = NOT ( ( fin1 ) )
cdc42 = ( ( scd1 ) AND NOT ( Rga4 ) ) OR ( ( gef1 ) AND NOT ( Rga4 ) )
pak1 = ( cdc42 )
orb6 = ( nak1 ) OR ( pak1 )
pmo25 = ( cdc14-sid1 ) OR ( cdc7 )
nak1 = ( sid2-mob1 AND ( ( ( cdk-H ) ) ) ) OR ( pmo25 )
cdc14-sid1 = ( cdc7 )
|
RESEARCH ARTICLE
A Dynamic Gene Regulatory Network Model
That Recovers the Cyclic Behavior of
Arabidopsis thaliana Cell Cycle
Elizabeth Ortiz-Gutiérrez1,2, Karla García-Cruz1, Eugenio Azpeitia1,2¤, Aaron Castillo1,2,
María de la Paz Sánchez1, Elena R. Álvarez-Buylla1,2*
1 Instituto de Ecología, Universidad Nacional Autónoma de México, 3er Circuito Exterior, Junto a Jardín
Botánico Exterior, México, D.F. CP 04510, México, 2 Centro de Ciencias de la Complejidad-C3, Universidad
Nacional Autónoma de México, Ciudad Universitaria, Apartado Postal 70–275, México, D.F. 04510, México
¤ INRIA project-team Virtual Plants, joint with CIRAD and INRA, UMR AGAP, Montpellier, France
* eabuylla@gmail.com
Abstract
Cell cycle control is fundamental in eukaryotic development. Several modeling efforts have
been used to integrate the complex network of interacting molecular components involved
in cell cycle dynamics. In this paper, we aimed at recovering the regulatory logic upstream
of previously known components of cell cycle control, with the aim of understanding the
mechanisms underlying the emergence of the cyclic behavior of such components. We
focus on Arabidopsis thaliana, but given that many components of cell cycle regulation are
conserved among eukaryotes, when experimental data for this system was not available,
we considered experimental results from yeast and animal systems. We are proposing a
Boolean gene regulatory network (GRN) that converges into only one robust limit cycle
attractor that closely resembles the cyclic behavior of the key cell-cycle molecular compo-
nents and other regulators considered here. We validate the model by comparing our in sil-
ico configurations with data from loss- and gain-of-function mutants, where the endocyclic
behavior also was recovered. Additionally, we approximate a continuous model and recov-
ered the temporal periodic expression profiles of the cell-cycle molecular components
involved, thus suggesting that the single limit cycle attractor recovered with the Boolean
model is not an artifact of its discrete and synchronous nature, but rather an emergent con-
sequence of the inherent characteristics of the regulatory logic proposed here. This dynam-
ical model, hence provides a novel theoretical framework to address cell cycle regulation in
plants, and it can also be used to propose novel predictions regarding cell cycle regulation
in other eukaryotes.
Author Summary
In multicellular organisms, cells undergo a cyclic behavior of DNA duplication and deliv-
ery of a copy to daughter cells during cell division. In each of the main cell-cycle (CC)
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
1 / 28
a11111
OPEN ACCESS
Citation: Ortiz-Gutiérrez E, García-Cruz K, Azpeitia
E, Castillo A, Sánchez MdlP, Álvarez-Buylla ER
(2015) A Dynamic Gene Regulatory Network Model
That Recovers the Cyclic Behavior of Arabidopsis
thaliana Cell Cycle. PLoS Comput Biol 11(9):
e1004486. doi:10.1371/journal.pcbi.1004486
Editor: Reka Albert, UNITED STATES
Received: February 27, 2015
Accepted: August 3, 2015
Published: September 4, 2015
Copyright: © 2015 Ortiz-Gutiérrez et al. This is an
open access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Funding: This study was financed with the following
grants: CONACyT:180098 and 180380 (ERAB),
152649 (MPS); and UNAM-DGAPA-PAPIIT:
IN203113 (ERAB) and IN203814 (MPS). The funders
had no role in study design, data collection and
analysis, decision to publish, or preparation of the
manuscript.
Competing Interests: The authors have declared
that no competing interests exist.
stages different sets of proteins are active and genes are expressed. Understanding how
such cycling cellular behavior emerges and is robustly maintained in the face of changing
developmental and environmental conditions, remains a fundamental challenge of biol-
ogy. The molecular components that cycle through DNA duplication and citokinesis are
interconnected in a complex regulatory network. Several models of such network have
been proposed, although the regulatory network that robustly recovers a limit-cycle steady
state that resembles the behavior of CC molecular components has been recovered only in
a few cases, and no comprehensive model exists for plants. In this paper we used the plant
Arabidopsis thaliana, as a study system to propose a core regulatory network to recover a
cyclic attractor that mimics the oscillatory behavior of the key CC components. Our analy-
ses show that the proposed GRN model is robust to transient alterations, and is validated
with the loss- and gain-of-function mutants of the CC components. The interactions pro-
posed for Arabidopsis thaliana CC can inspire predictions for further uncovering regula-
tory motifs in the CC of other organisms including human.
Introduction
The eukaryotic cell cycle (CC) in multicellular organisms is regulated spatio-temporally to
yield normal morphogenetic patterns. In plants, organogenesis occurs over the entire lifespan,
thus CC arrest, reactivation, and cell differentiation, as well as endoreduplication should be
dynamically controlled at different points in time and space [1]. Endoreduplication is a varia-
tion of the CC, in which cells increase their ploidy but do not divide. Normal morphogenesis
thus depends on a tight molecular coordination among cell proliferation, cell differentiation,
cell death and quiescence. These biological processes share common regulators which are influ-
enced by environmental and developmental stimuli [1–3]. It would not be parsimonious to
depend on different regulatory circuits to control such interlinked cellular processes, CC
behaviors and responses. Thus we postulate that a common network is deployed in all of them.
Such overall conserved CC network may then connect to different regulatory networks under-
lying cell differentiation in contrasting tissue types or to signal transduction pathways elicited
under different conditions, and thus yield the emergence of contrasting cellular behaviors in
terms of cycling rate, entrance to endocycle, differentiation, etc.
Furthermore, the overall CC behaviors are widely conserved and robust among plants and
animals. Hence, we aim at further investigating the collective behavior of the key upstream reg-
ulators and studied CC components to understand the mechanisms involved in the robustness
of CC regulation under changing developmental stages and environmental conditions faced by
plants along their life-cycles. Previous studies, that have shown the oscillatory behavior of sev-
eral transcription factors, that had not been associated as direct regulators of the CC, support
our proposed hypothesis [4]. We thus propose to uncovering the set of necessary and sufficient
regulatory interactions underlying the core regulatory network of plant CC, including some
key upstream transcriptional regulators.
Computational tools are essential to understanding the collective and dynamical behavior
of these components within the regulatory networks involved. As a means of uncovering the
main topological and architectural traits of such networks, we propose to use Boolean formal-
isms that are simple and have proven to be useful and powerful to follow changes in the activity
of regulators of complex networks in different organisms and biological processes [5, 6].
Although the key CC components have been described in different organisms, the complex-
ity and dynamic nature of the molecular interactions that are involved in CC regulation and
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
2 / 28
the emergence of the cyclic behavior of the CC molecular components are not well understood
yet. The use of systemic, dynamic and mathematical or computational approaches has been
useful towards this already. Previous models have focused mainly on yeast and animal systems
and have been useful to analyze many traits of CC behavior such as robustness, hysteresis, irre-
versibility and bistability [7–11]. The latter two properties have been validated with experimen-
tal data [12–14].
We herein summarize the main traits and components of the eukaryotic CC. The molecular
CC regulators have been described and they are well conserved across distantly related organ-
isms [15, 16]. CC progression is regulated by Cyclin-Dependent Kinases (CDKs) [17] that asso-
ciate with different cyclins to confer substrate specificity [18]. CDK-cyclin complexes trigger
the transition from G1 (Gap 1) to synthesis phase (S phase) in where the genome is duplicated,
and from G2 (Gap 2) to mitotic phase (M phase) for the delivery of the newly duplicated DNA
to the two daughter cells [19] (see for a review [17, 20]). The CDK-cyclin activity also regulates
the cell transit between G and S phases during the endoreduplication process [21, 22].
Two CDKs (CDKA and CDKB) are involved in CC regulation. CDKA;1-CYCDs and
CDKA;1-CYCA3 complexes regulate G1/S and S phase progression [23–25]; while
CDKB-CYCA2 and CDKB-CYCBs regulate G2/M phase and M progression [26–28]. Thus
CDK-cyclin activity is finely-tuned by phosphorylation, interactions with CDK inhibitors such
as Kip-related proteins (KRPs), and degradation of cyclins and KRPs by Skp1/Cullin/F-
box (SCF), as well as by the anaphase-promoting complex/Cyclosome (APC/C) [29–31].
Besides these components, plant CC machinery has a greater number of CC regulators than
other eukaryotes and some of those components such as the CDKB are plant-specific.
Several key transcriptional regulators participate in the G1/S and G2/M transitions [32].
The E2F/RBR pathway regulates G1/S transition by transcriptional modulation of many genes
required for CC progression and DNA replication [33, 34]. While E2Fa and E2Fb with their
dimerization partner (DP) activate transcription of a subset of S phase genes, E2Fc-DP
represses transcription [35]. The function of E2Fa and E2Fb is inhibited by their interaction
with RBR [36]; in G1/S transition CDKA;1-CYCD-mediated RBR hyperphosphorylation,
releases E2Fa/b-DP heterodimers allowing transcriptional activation of E2Fa and E2Fb targets.
Simultaneously the E2Fc-DP transcriptional inhibitor is degraded [37].
Little is known about the regulation of G2/M transition in plants, however a class of con-
served transcription factors belonging to the MYB family has been described, that seem to have
key roles in CC regulation. MYB transcription factors have a prominent role during G2/M
transition, by regulating, for example, CYCB1;1 which is determinant in triggering mitosis
[38–43]. For the mitosis exit, APC/C mediates degradation of the mitotic cyclins as CYCB1;1
and CYCA2;3, inactivating CDK-cyclin complexes. CCS52A2, an activator subunit of APC/C,
is transcriptionally inhibited by E2Fe [44].
Some previous models have recovered the limit cycle attractor as well for CC components
[45–48]. A pioneer model of the CC focused on mitotic CDK-cyclin heterodimer and a cyclin
protease oscillatory behavior [49]. On the other hand, Novak and Tyson incorporated addi-
tional nodes and interactions to model the G1/S and G2/M transitions of the S. pombe CC [50,
51]. They also analyzed evolutionary roles of CC regulators [52], mutant phenotypes [53], sta-
ble steady states [7] and the role of cues such as cell size or pheromones in CC progression [54,
55]. Additionally, comprehensive CC continuous models [45] and generic modules for eukary-
otic CC regulation [56, 57] have been proposed.
In addition to continuous formalisms, CC models have used discrete approaches as Boolean
models for yeast and mammalian systems [46–48, 58–61], and more recently, hybrid models
for mammalian cells have been published [62]. Subsequently, time-delayed variables [63] and
variables defining CC events [47, 48] were incorporated. Time robustness was improved with
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
3 / 28
specifications of the temporal order with which each component is activated [60]. Recent pub-
lished reports on CC dynamics use steady state probability distributions and potential land-
scapes, and highlight the enormous potential of CC models to characterize normal and altered
regulation of mammalian CC [64, 65].
Yeast CC Boolean models with summatory thresholds [58, 59], incorporated self-degrada-
tion for proteins, but did not incorporate several negative regulators explicitly. In a later work
[61], nodes were kept active when the summatory effect of their regulators was greater than the
activation threshold, which implies self-degradation of the protein, when such summatory is
equal to or below the threshold. Fauré and Thieffry have transformed CC Boolean models, that
use threshold functions, to models with a combinatorial scheme, and they have also presented
a broader discussion about these two approaches to logical frameworks [66].
Two Boolean models of budding yeast CC and another one of mammalian CC recover cyclic
attractors [46–48]. The mammalian CC model [46] also recovers a fixed-point attractor corre-
sponding to G0. In another study, Fauré and collaborators integrated three modules to yield a
comprehensive model for the budding yeast CC GRN [47]. The components included variables
to represent cellular growth, citokinesis, bud formation, DNA replication and the formation of
the spindle. The yeast CC model by Irons also included variables of CC events (e.g. bud forma-
tion or DNA replication) as well as time delays [48]. In contrast to other eukaryotes, in Arabi-
dopsis thaliana (A. thaliana herein) very few attempts have been made to integrate available
experimental data on CC regulators using mechanistic models. Only a study that considers the
G1/S transition has been proposed and contributed to show some additional conserved features
of this CC control point among eukaryotes [67].
We integrated available experimental data on 29 A. thaliana regulatory interactions involved
in CC progression into a Boolean discrete model, that recovers key properties of the observed
plant CC. The regulatory network, that we put forward, also incorporates three uncovered inter-
actions, based on animal systems (E2Fb ! SCF, CDKB1;1-CYCA2;3 a E2Fa, APC/C a SCF), as
well as 16 interactions based on bioinformatic analyses. Therefore, the latter proposed interac-
tions constitute new predictions that should be tested experimentally. The use of yeast or animal
data is supported by the fact that main CC components or regulatory motifs are conserved
among eukaryotes [16]. In our model, we include solely molecular components and avoid artifi-
cial self-degradation loops, which have been used for recovering the limit cycle attractor. We vali-
dated the model simulating loss- and gain-of-function lines, and hence demonstrate that the
Boolean network robustly implements true dynamical features of the biological CC regulatory
network under wild type and genetic alterations. Possible artifacts due to the discrete dynamical
nature of the model used, and of its synchronous updating scheme, were discarded by comparing
the Boolean model results to those of a continuous approximation model. The continuous model
indeed recovers the robust limit cycle that mimics the dynamical behavior of CC components
under a wide range of parameters tested. Finally, we provide novel predictions that can be tested
against biological experimental measurements in future studies. The model put forward consti-
tutes a first mechanistic and integrative explanation to A. thaliana CC.
Materials and Methods
Boolean model
We proposed a Boolean approach to integrate and study the qualitative complex logic of regu-
lation of the molecular components underlying the CC dynamics. We formalized available
experimental data on logical functions and tables of truth that rule how the state of a particular
component is altered as a function of the states of all the components that regulate it. In a Bool-
ean model each node state can be 0, when the expression of a gene or other type of molecular
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
4 / 28
component or complex of such components is unexpressed or “OFF”, or 1 when it is expressed,
or “ON”. Nodes states are updated according to the function: Xi(t+1) = Fi(Xi1(t), Xi2(t), . . .,
Xik(t)), where Xi(t+1) is the state of Xi gene at time t+1 and Xi1(t), Xi2(t), . . ., Xik(t) is the set of
its regulators at time t. The set of logical rules for all the network components defines the
dynamics of the system. By applying the logical rules to all nodes for several iterations, the
dynamics of the whole network can be followed until it reaches a steady state; a configuration
or set of configurations that does not change any more or are visited in a cyclical manner,
respectively. Such state is called an “attractor”. Single-point attractors only have one GRN con-
figuration, or cyclic attractors with period n, which have n configurations that are visited indef-
initely in the same order. In this paper we propose a GRN model that converges to a single
limit cycle attractor that recovers the CC molecular components’ states of presence (network
configuration) in a cyclic pattern that mimics the pattern observed for the molecular compo-
nents included in the model along the different CC phase.
Model assumptions
A. thaliana CC Boolean model has the following assumptions:
1. Nodes represent mRNA, proteins or protein complexes involved in CC phase transitions.
Node state “ON” is for the presence of regulator, and “OFF” is for absence; in the latter case,
it may also indicate instances in which a component may be present but non-functional due
to a post-translational modification.
2. The state of the RBR (RETINOBLASTOMA-RELATED) node corresponds to a 1 or “ON”
when this protein is in its hypo-phosphorylated form and therefore is ready to inhibit E2F
transcription factors.
3. When a particular CDK is not specified, a cyclin can form a complex with CDKA;1, a kinase
that is always present because it is expressed in proliferative tissues [68] during the complete
CC.
4. E2Fa, E2Fb and E2Fc need dimerization partner proteins (DPa or DPb) for its DNA-bind-
ing. Given that DP expression does not change drastically in CC [69], we assumed that the
state of these heterodimers is given only by the presence of E2F factors.
5. The Boolean logical functions integrate and formalize experimental data available mainly
for the A. thaliana root apical meristem, however some data from leaves were considered,
and we assumed that these are also valid for CC regulation in the root meristem. Also, data
from other systems and data obtained by sequence promoter analysis were considered as
indicated in each case [27, 39, 40, 67, 70–85] (summarized in Table 1).
6. The dynamics of complex formation (such as CDK-cyclin and KRP1, or RBR and E2F fac-
tors) are specified directly in the Boolean function of their target genes. For instance, the
logic rule for E2Fb is E2Fa & !RBR, indicating that E2Fb state is “ON” when it is transcrip-
tionally activated by E2Fa free of RBR. All E2Fa targets also included in their logical rules
RBR, as is shown in S1 Text. Then, the presence of KRP1 or RBR in a logical rule does not
imply that they are regulators acting directly on the corresponding target.
7. The updating scheme for the node states was synchronous.
Periodic expression and promoter sequence analysis
Most regulatory interactions and logical rules were obtained from the A. thaliana data [20, 21,
25–27, 29, 30, 35, 37, 38, 40, 43, 44, 78–80, 85–103] (detailed in Table 2). A. thaliana CC-
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
5 / 28
dependent expression data for validation was obtained from: [72–74]. The consensus site used
for MYB77 was CNGTTR, according to: [75, 76], while that for MYB3R4 was AACGG accord-
ing to: [43]. The motifs were searched in the regulatory sequences of all network nodes using
Pathmatch tool (http://arabidopsis.org/cgi-bin/patmatch/nph-patmatch.pl) of TAIR. Regula-
tory sequences in TAIR10 Loci Upstream Sequences-1000bp and TAIR10 5’ UTRs datasets
were used.
Software for robustness analysis and mutant simulation
We used BoolNet [104] (a library of R language [105]) and Atalia(Á. Chaos; http://web.
ecologia.unam.mx/achaos/Atalia/atalia.htm) to simulate the CC GRN dynamics and perform
robustness, and mutant analyses. Systematic alterations in Boolean functions for robustness
Table 1. Hypothetical Interactions for the A. thaliana CC Network.
Regulator
Target
Data supporting the proposition of the interaction
Refs.
E2Fb
!
SCF
F-box protein Skp2 is part of the SCF complex and is transcriptionally
regulated by E2F1 in humans. In A. thaliana, it has only been reported that
E2F factors regulate FBL17, another F-box protein.
[67, 70]
E2Fb
!
MYB77
Direct regulation between E2F and MYB factors has been reported in
budding yeast and mammals, but in plants it could include at least one
intermediary; A. thaliana could have a similar regulation because its CC also
presents transcriptional waves in G1/S and G2/M transitions as yeasts and
mammals. After analyzing the two main families of transcription factors
involved in CC regulation: TCP and MYB, we propose MYB77 as a mediator
between E2F and MYB regulation. Using available microarray analyses, we
found that MYB77 shows CC-dependent expression with a peak in M phase.
In addition to having binding sites for E2F, with the identification of the
binding site recognized by MYB77, we can hypothesize that MYB77
regulates MYB3R1/4 and other CC genes.
[39,
71–74]
MYB77
!
E2Fe, KRP1, MYB3R1/4,
CYCB1;1, CYCA2;3, CDKB1;1,
CCS52A2
The sequence CNGTTR identified as a consensus site recognized by
MYB77 was used to find its possible targets among CC core genes. Several
of them are expressed just before G2 to M phase transition.
[75–77]
MYB3R1/4
!
SCF, RBR, CDKB1;1, CYCA2;3,
APC/C, E2Fc, MYB3R1/4, KRP1
The consensus site of MYB3R4 was found in SKP2A, RBR, CDKB1;1,
CYCA2;3, CCS52A2, KRP1, E2Fc, MYB3R1/4 and CYCB1;1 by in silico
analysis described in the Materials and Methods section. In tobacco,
NtmybA1 and NtmybA2 genes have the MSA sequence and they can
regulate themselves. MYB3R1/4 might promote the expression of KRP1,
since KRP1 has a peak of expression in G2/M and has eight putative MSA
elements. CYCB1;1 regulation by MYB3R1/4 also has experimental support.
[40, 78]
CDKB1;1-CYCA2;3
a
E2Fa
It has been hypothesized that a cause of low levels of E2Fa could be due to
its high turnover rate as result of CDKB1;1 phosphorylation. This E2F factor
has putative CDK-phosphorylation sites in its N-terminal end, and a high
CDK activity inversely correlates with its DNA binding ability in vitro. This
hypothesis is supported by observations in mammalian cells.
[27,
79–81]
APC/C
a
SCF
It was proposed that APC/C and SCF functions are mutually exclusive during
CC progression, which led to the identification of the relationship amongst
them. In proliferating mammal cells, levels of Skp2, a SCF subunit, oscillate
under the pattern of APC/C substrates. Furthermore, the APC/C subunit
Cdh1 participates in the degradation of Skp2 and the reduction of Cdh1
expression stabilizes Skp2. A. thaliana SCF and APC/C seem have the same
roles during CC as their animal counterparts.
[82–85]
A summary of the data led us to propose interactions that have not been previously described for A. thaliana CC. a stands for negative regulation and !
for positive regulation.
doi:10.1371/journal.pcbi.1004486.t001
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
6 / 28
analyses were done with Atalia, while stochastic perturbations in random networks to compare
attractor’s robustness were done with BoolNet. For random perturbations made in transitions
between network configurations or in Boolean functions, the “bitflip” method was applied. To
validate the GRN model proposed here, we used BoolNet and simulated loss- and gain-of-func-
tion mutations for each node, by skipping the node’s logical rule and setting the respective
gene to “0” and “1”, respectively.
Table 2. Experimental Interactions for the A. thaliana CC Network and their Evidence.
Regulator
Target
Description of the interaction
Refs.
CDKA;1-CYCD3;1
a
RBR
Studies suggest that complexes formed by CDKA;1 and D-type cyclins phosphorylate RBR.
[20, 86–89]
CDKA;1-CYCD3;1
a
RBR–
E2Fb
E2Fb–RBR complex diminishes in CYCD3;1 overexpressor line.
[90]
CDKA;1-CYCD3;1
a
E2Fc
CDKA;1 bound to D-type cyclin affects formation of E2Fc-DPb complex and its binding to DNA.
The recognition of E2Fc by the SCF complex depends on phosphorylation mediated by CDKA;1.
[35, 37, 91]
SCF
a
CYCD3;1
SCF is involved in the ubiquitination required for CYCD3;1 degradation.
[92]
SCF
a
KRP1
SCF ubiquitinates KRP1 to be degraded.
[85, 93]
SCF
a
E2Fc
E2Fc shows the accumulation in skp2a mutant (subunit of SCF); the overexpression of SKP2A
reduces levels of E2Fc.
[35, 91]
RBR
a
E2Fa/b
RBR is a negative regulator of E2Fa/b transcriptional activity.
[90]
E2Fa
!
E2Fb
E2Fb transcription is induced in E2Fa overexpressor line.
[94]
E2Fa
!
E2Fc
E2Fc has binding sites for E2F and it is induced in E2Fa-DPa overexpressors.
[80, 94]
E2Fa
!
RBR
Transcriptional control of RBR is under E2Fa transcriptional activity.
[95]
E2Fa
!
APC/C
CCS52A2, a component of APC/C, is induced when RBR-free E2Fa is overexpressed.
[90]
E2Fb
!
CYCB1;1
CYCB1;1 expression is induced when RBR-free E2Fb increases; targets of E2Fb are genes
needed for G2/M transition.
[79, 80, 90]
E2Fb
!
CDKB1;1
Inducible expression of E2Fb promotes CDKB1;1 expression.
[79]
E2Fb
!
E2Fe
E2Fb induces transcription of E2Fe.
[96]
E2Fc
a
CDKB1;1
The effect of E2Fb can be countered by E2Fc; with E2Fc destabilization increments CDKB1;1.
[96, 97]
E2Fc
a
CYCB1;1
CYCB1;1 expression increases when E2Fc expression is silenced; E2Fc overexpression reduces
CYCB1;1 level.
[37]
E2Fc
a
E2Fa
E2Fa messengers increase when E2Fc expression is silenced.
[37]
E2Fc
a
E2Fe
E2Fc counteracts the positive effect that E2Fb has in the expression of E2Fe.
[96]
E2Fe
a
APC/C
Expression of CCS52A, a subunit of APC/C, is downregulated by E2Fe.
[44]
MYB3R1/4
!
CYCB1;1
MYB3R1/4 recognizes the sequence AACGG required for CYCB1;1 expression; other regulators
seem to drive its G2/M-specific expression.
[38, 43]
CDKB1;1
–
CYCA2;3
CYCA2;3 interacts with CDKB1;1 to form a functional complex.
[25, 27]
CDKB1;1-CYCA2;3
a
KRP1
In complex with CYCA2;3, CDKB1;1 could promote KRP1 proteolysis as promotes KRP2
proteolysis; both KRPs could have similar roles in mitosis entry, since both interact with CDKA;1
and are expressed in G2/M.
[21, 27, 78]
CDKB1;1, CDKA;1
–
CYCB1;1
B-type cyclins interact with B-type and A-type CDKs.
[25, 26]
CDKA;1-CYCB1;1
!
MYB3R1/
4
The overexpression of MYB3R4 enhances the 2-fold activity of its target promoters in comparison
to WT, and the co-expression of MYB3R4 and CYCB1;1 enhances them 4-fold; CycB1 and other
mitotic cyclins enhances the activity of NtmybA2 factors in tobacco.
[40, 98, 99]
KRP1
a
CYCD3;1
KRP1 is able to interact with CDKA;1 and CYCD3;1.
[29, 93, 100,
101]
KRP1
a
CYCB1;1
KRP1 binding to CDKA;1 inhibits the activity of CDKA–CYCB1;1.
[30, 100]
APC/C
a
CYCB1;1
The APC/C complex ubiquitinates CYCB1;1 to be degraded.
[102]
APC/C
a
CYCA2;3
CYCA2;3 is stabilized with loss-of-function mutations in APC/C subunits or with mutations in its D-
box.
[27, 103]
Summary of experimental evidence supporting interactions of A. thaliana CC GRN. a represents negative regulation, ! is for positive and — represents
the formation of functional complex.
doi:10.1371/journal.pcbi.1004486.t002
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
7 / 28
Continuous model
For the continuous model, we followed [106, 107]. In the continuous version of the model the
rate of change for each xi node is represented by a differential equation that comprises produc-
tion as well as decay rates:
dxi
dt ¼
e0:5h þ ehðoiÞ
ð1 e0:5hÞ ð1 þ ehðoi0:5ÞÞ gixi
ð1Þ
The parameter h determines the form of the curve; when h is very close to 0, the curve
becomes a straight line, while with values close to 100, the curve approximates a step function.
The parameter ωi is the continuous form of Fi(Xi1(t), Xi2(t), . . ., Xik(t)) used in the Boolean
model, and γi is its degradation rate. Detailed information about the continuous model can be
found in S2 Text.
Results
The regulatory network recovers a dynamical model of A. thaliana CC
The CC model proposed here integrates and synthesizes published data for A. thaliana CC
components interactions, as well as some molecular data from other organisms (mammal and
yeast), that we propose as predictions for A. thaliana CC regulation, and assume to be con-
served among all eukaryotes. The whole set of interactions and nodes included in the model
and detailed in Tables 1 and 2 are shown in Fig 1. Four types of molecular interactions can be
distinguished: (i) transcriptional regulation, (ii) ubiquitination, (iii) phosphorylation and (iv)
physical protein-protein interactions. Additionally, an in silico analysis of transcription factors
and promoters was carried out, in order to further substantiate 16 predicted interactions in the
GRN (these are: E2Fb ! MYB77; MYB77 ! E2Fe, MYB3R1/4, KRP1, CYCB1;1, CYCA2;3,
CDKB1;1 and CCS52A2; MYB3R1/4 ! SCF, RBR, CDKB1;1, CYCA2;3, APC/C, KRP1, E2Fc
and MYB3R1/4). The logical rules are available in S1 Text.
Our results show that the nodes and interactions considered are sufficient to recover a single
robust cyclic steady state, and thus the cyclic behavior of the components considered. Such
behavior closely resembles the periodic patterns observed during actual CC progression, Fig 2.
The first two columns or network configurations match a G1 state, given that during the early
G1 phase, the CDKA;1-CYCD3;1 complex is absent or inactive by the presence of KRP1 [92,
93, 108]. The CDKA;1-CYCD3;1 state is given only by the presence of CYCD3;1 since CDKA;1
is always expressed in proliferative cells [68]. To facilitate understanding, in Fig 2 the complex
CDKA;1-CYCD3;1 is shown instead of only CYCD3;1. The absence of mitotic cyclins
(CYCA2;3 and CYCB1;1) at this stage [28, 38], as well as the APC/C presence until the early
G1 phase, which is needed for the mitosis exit, also coincides with experimental observations
[44, 109, 110]. The presence of the RBR protein in G1-phase implies an inactive state of the
E2F, as expected [33, 111, 112]. Then, the third column resembles G1/S transition, where the
presence of CDKA;1-CYCD3;1 complex would be inducing RBR phosphorylation and its inac-
tivation [32]. In the fourth configuration, the S-phase is represented by RBR inactivation and
E2Fa/b transcriptional activation [113]. In the fifth and sixth configuration, E2Fc state returns
to “ON” but RBR state is kept in “OFF”, which indicates that transcription driven by E2Fa and
E2Fb can still happen. Indeed, the E2Fb factor appears from the fifth configuration and it is
consistent with their function regulating the expression of genes needed to achieve the G2/M
transition. In the sixth configuration, MYB77 is turned on, although in synchronization experi-
ments it has been observed to be on until the beginning of mitosis [73]. During G2-phase the
MYB transcription factors and KRP1 are expressed [31, 73, 93], the former would maintain
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
8 / 28
dimers of CDKA;1 and mitotic cyclins inactive; and together, this data is consistent with what
is observed in the seventh configuration of the CC attractor. In the eighth column, KRP1 is lost
because it was phosphorylated by CDKB1;1-CYCA2;3, which is active in the G2/M transition
and the onset of mitosis [27]. The phosphorylation of KRP1 drives its degradation and poste-
rior activation of mitotic complexes such as CDKA;1-CYCB1;1 to trigger mitosis [21, 78] (con-
figuration 9 and 10 in Fig 2). The lack of APC/C at the onset of mitosis is determinant for the
accumulation of the mitotic cyclins, but APC/C presence is necessary for the mitosis exit [110],
which occurs in the eleventh configuration of the attractor (Fig 2). Thus, our CC GRN model
recovers a unique attractor of eleven network configurations (Fig 2), which shows a congruent
Fig 1. Regulatory network of the A. thaliana CC. The network topology depicts the proteins included in the model as well as the relationship among them.
Nodes are proteins or complexes of proteins and edges stand for the existing types of relationships among nodes. The trapezoid nodes are transcription
factors, the circles are cyclins, the squares are CDKs, the triangle represent stoichiometric CDK inhibitor, the hexagons are E3-ubiquitin ligase complexes
and the octagon is a negative regulator of E2F proteins. Edges with arrow heads are positive regulations and edges with flat ends illustrate negative
regulations. The red edges indicate regulation by phosphorylation while blue ones indicate ubiquitination, the green ones show physical protein-protein
interactions and the black edges transcriptional regulation. Only CDK-cyclin interactions are not represented with a line. Interactions to or from rhombuses
stand for interactions that involve the CDK as well as the cyclin. A solid line indicates that there is experimental evidence to support such interaction and
dotted lines represent proposed interactions grounded on evidence from other organisms or in silico analysis.
doi:10.1371/journal.pcbi.1004486.g001
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
9 / 28
cyclic behavior of its components with that observed experimentally. This result validates that
the proposed set of restrictions converge to a single cyclic behavior, which is independent of
the initial conditions. A further validation of the proposed CC model, would imply that the
recovered cyclic attractor is robust to permanent alterations, as is the case for real CC behavior
that is highly robust to external and internal perturbations [14, 58, 114, 115].
The CC Boolean model is robust to alterations
To provide further validation for the proposed CC regulatory network, we performed robust-
ness analyses of the attractor to four types of alterations in the logical functions of the model.
First, we altered the output of each logical rule by systematically flipping one by one, each one
of their bits. We found that 87.47% of the perturbed networks recovered the original attractor,
while 1.77% of the altered networks maintained the original attractor and produced new ones
(see supplementary material S3 Text for details). In contrast, the remaining 10.76% of alter-
ations reduced the number of network configurations of the original attractor. In the second
robustness analysis, after calculating the transitions between one network configuration to the
next one, one bit (i.e. the state of a node) of this next configuration is randomly chosen and its
value changed. Then, the network is reconstructed and its attractors recovered again. This pro-
cedure was repeated 100 times, thus we found that in 88.2 ± 3.2 out of the 100 perturbations
(mean ± SD) the original attractor was reached. These results suggest that the proposed GRN
for A. thaliana CC is robust to alterations as expected and in coincidence with previous GRN
models proposed for other developmental processes [116, 117].
To confirm that the robustness recovered in these two types of analyses is a specific property
of the network under study, we performed robustness analyses of randomly generated
Fig 2. Attractor corresponding to a dynamic network of CC in A. thaliana. 100% of the whole set of network configurations converges to a unique
attractor composed by 11 configurations. Each column is a network configuration (state of each network component) and the rows represent the state of each
node during CC progression. The squares in green indicate components that are in an “ON” state and the ones in red are nodes in an “OFF” state.
doi:10.1371/journal.pcbi.1004486.g002
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
10 / 28
networks with similar structures (same number of input interactors for the logical functions)
to the one proposed here for the A. thaliana CC regulatory network, and compared the above
robustness analyses results to those recovered for equivalent analyses for the random networks.
We generated 1000 random networks. Then, 100 copies of the random and of our network
were done. In each copy we randomly flipped the value of one bit in one logical function (to
confirm the first robustness analysis), or in one next configuration (for the second robustness
analysis). When perturbations are made in logical functions, the A. thaliana CC GRN recovers
its attractor in 68% of perturbations, while the median of percentage of cases in which such
attractor was recovered in the random networks was only 18.55% (mean 19.12% ± 13.86 SD,
Fig 3A). The difference between the 68% of this latter analysis and the 87.47% of the first
robustness analysis could be due to sampling error. If transitions between network configura-
tions are perturbed, the median of original attractors recovered in random networks is 24.2%
(mean 24.6% ± 18.2 SD). In contrast, the original attractor of A. thaliana CC GRN was found
in 88% of perturbed networks starting with that grounded on experimental data (Fig 3B).
These results confirm that the CC GRN proposed here is much more robust than randomly
generated networks with similar topologies and suggests that its robustness is not due to overall
structural properties of the network.
Boolean models can produce cyclic dynamics as an artifact due to their discrete nature and
the time delays implied. To address this issue we approximated the Boolean model to a contin-
uous system of differential equations following [106, 107, 118, 119]. To recover steady states of
such continuous system, the continuous versions of the GRN were evaluated for 1000 different
randomly picked initial conditions (See S2 Text). In all cases and independently of the method-
ology (i.e. [106, 107] or [118, 119]), we recovered the same limit cycle steady state. In the con-
tinuous model, key cyclins for the main phase transitions, CYCD3;1 and CYCB1;1, have an
oscillatory behavior that is not attenuated with time (Fig 4). Importantly, this result is robust to
changes in the decay rates or alterations of the h parameter that affects the shape of activation
function (see details in S2 Text); the limit cycle was recovered in 92.86% of the cases. The
results of the continuous model corroborate that the limit cycle attractor recovered by the Bool-
ean version, is not due to an artifact associated to the discrete and synchronous nature of the
Boolean model, but is rather an emergent property of the underlying network architecture and
topology. In addition, the recovery of the cyclic behavior of the continuous model constitutes a
further robustness test for the Boolean model.
Previous studies have also tested asynchronous updating schemes [46]. In this study we have
used a continuous form of the model to discard that the recovered cyclic attractor is due to an
artifact owing to the discrete and synchronous nature of the model used. Future studies could
approach analyses of asynchronous behavior of the model by devising some priority classes dis-
tinguishing fast and slow processes, and thus refining the asynchronous attractor, under a plausi-
ble updating scheme. On the other hand, biological time delays may be involved in CC
progression, but they are not enough for irreversibility. The CC unidirectionality has been pro-
posed to be a consequence of system-level regulation [120], here we hypothesize that the ordered
transitions of A. thaliana CC are an emergent property of network architecture and dynamics.
Simulated loss- and gain-of-function mutants recover observed patterns:
normal CC and endocycle
An additional validation analysis for the proposed A. thaliana CC model implies simulating
loss- and gain-of-function mutations and comparing the recovered attractors with the expres-
sion profiles documented experimentally for each mutant tested. We simulated mutants by fix-
ing the corresponding node to 0 or 1 in loss- and gain-of-functions mutations, respectively.
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
11 / 28
Fig 3. Attractor robustness analysis. Random networks with similar structure to A. thaliana CC GRN were
less tolerant to perturbations than original CC GRN. The frequency of perturbations that recovered the
original attractor after a perturbation in the Boolean functions, is shown in: (A), where the red line indicates
that A. thaliana CC GRN recovers its original attractor in 68% of perturbations (the median of random
networks was 18.55% and mean 19.12% ± 13.86 SD). When transitions between network configurations are
perturbed (B), A. thaliana CC GRN recovers its original attractor in 88% (vertical red line) of perturbations,
while the median of random networks that recover the original attractor was 24.2% (mean 24.6% ± 18.2 SD).
Vertical blue line indicates the 95% quantile. 1000 random networks were analyzed.
doi:10.1371/journal.pcbi.1004486.g003
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
12 / 28
The recovered altered configurations are summarized in S4 Text, and in Table 3 as well as in
Table 4 for gain- and loss-of-function mutants, respectively. The simulated mutant attractors
are coherent with experimental data in most cases [2, 21, 23, 30, 35, 37, 43, 44, 76, 79, 80, 88,
90–93, 103, 108, 109, 111, 113, 114, 121–129]. In Fig 5 we show a representative example of
attractors recovered by simulations of CDKB1;1 and KRP1 loss-of-function and APC/C and
E2Fa gain-of-function mutants. It is noteworthy that several simulated mutants, such as
mitotic cyclins or B-type CDK loss-of-function, converge to a cyclic attractor that corresponds
to the configuration observed under an endoreduplicative cycle (e.g. Fig 5A). In such attractors,
endoreduplication inductors, such as APC/C, KRP1 and E2Fc [37, 78, 130] are present, at least
in some network configurations (Fig 5A, 5C and 5D-right). Another outstanding feature of
these mutant attractors is that, although mitotic CDK-cyclin complex may be present, it is
inhibited by KRP1, therefore there is no CDK-cyclin activity to trigger the onset of mitosis.
These data are coincident with the reported regulation during the onset of endoreduplication
[21]. In the attractors where E2Fa coincides with alternating states of RBR, it suggests that
DNA replication may occur (Fig 5). Likely due to plant redundancy, some mutations do not
produce an obvious impaired phenotype. Such is the case of KRP1 loss-of-function, in which
loss-of-function simulation, a cyclic attractor identical to the original one is recovered, as is
expected (see Table 4), because such mutants do not show an evident altered CC behavior (Fig
5B) [93].
Interestingly, the simulation of a constitutively active APC/C also converges to a single
cyclic attractor, which corresponds to an endoreduplication cycle, since it has Gap and S
phases, but lacks an M-phase configuration. This coincides with the experimental observation
that the overexpression of one of the APC/C subunits (CCS52A) promotes entry to an endo-
cycle [44] (see Table 3). Another interesting example is the gain-of-function mutation of E2Fa
that yields two cyclic attractors, one corresponding to the normal CC cycle and the other one
Fig 4. Continuous version of the A. thaliana CC Boolean model. In this graph we show the activity of the CDKA;1-CYCD3;1 and the CDKA;1-CYCB1;1
complexes as a function of the amount of cyclins, and KRP1 inhibitor. The CDK-cyclin activity is the limiting factor to pass the G1/S and the G2/M
checkpoints. A little more than two complete CC are shown (upper horizontal axis) to confirm that oscillations are maintained.
doi:10.1371/journal.pcbi.1004486.g004
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
13 / 28
to an endocycle (Table 3). It has been shown that this gene is required for both processes [111]
that are apparently exclusive, although in both processes the DNA replication occurs and
among E2Fa targets there are genes required for S-phase. Thus our model suggests that the reg-
ulation of E2Fa at the end of G2 phase is decisive for CC exit and transition to endoreduplica-
tion. In this E2Fa gain-of-function simulation, we found an inconsistency with APC/C because
this E3 ubiquitin ligase is decisive for endoreduplication, while in the simulated attractor is
only present in one network configuration (Fig 5D-right). Such behavior observed in the
endoreduplication attractor for E2Fa gain-of-function leads to unstable activity in the CDK-
cyclin complex (Fig 5D), thus suggesting that the increase in APC/C is required for endoredu-
plication entry as well as its progression. In the attractor of the simulated APC/C gain-of-func-
tion, the states of the CYCD3;1, SCF, E2Fb, E2Fc and MYB nodes are more stable than in
Table 3. Phenotypes of gain-of-function mutations in CC components.
Node
Phenotypes of gain of function
Recovered attractor(s)
Refs.
Model
CYCD3;1
Inhibition of CC exit, increases division zones and ectopic
divisions. Decreases G1 phase duration and increases G2
duration. Delays expression of G2/M genes.
Fixed-point attractor of G2-phase.
[108,
121]
PA
SCF
SKP2A gain-of-function enhances proliferation, and
increases number of cells in G2/M and ploidy levels
decrease.
Oscillates between G1 and S.
[122,
123]
NR
RBR
CC arrest, cells in root apical meristem lose CYCB1;1
expression; in rice, the number of cells synthesizing DNA
decrease.
Fixed-point attractor characterizing G1 arrest.
[2, 88]
A
E2Fa
Mitosis and endoreduplication are promoted.
One attractor comprising 40.48% of initial conditions that
is a WT CC. The other closely resembles an endocycle
but APC/C activity is lower (59.52% of configurations).
[111,
113]
A
E2Fb
Cell division is induced but endoreduplication is suppresed;
CC duration and cells are shorter, and the amount of S-
phase transcripts increases.
Similar to WT but with a shorter S phase.
[79, 80]
A
E2Fc
Overexpression of a non-degradable form of E2Fc leads to
larger cells or a lack of division.
Fixed-point attractor where only E2Fc and CYCD3;1 are
present, congruent with a CC-arrest.
[35]
PA
E2Fe
Reduces ploidy levels.
CC arrest in M phase.
[44]
PA
MYB77
Plants are stunted but there is no information about how CC
could be affected.
CC arrest in a mitotic state.
[76]
-
MYB3R1/
4
No available data about how it could alter cell division.
Two fixed-point attractors of CC arrest at early G1 phase,
state of E2Fa varies among them.
-
-
CYCB1;1
Root growth enhanced, slightly small cells.
WT CC
[124]
A
CDKB1;1
Does not seem to affect CC behavior.
WT CC
[125]
A
CYCA2;3
Not enough to produce multicellular trichomes but the
proportion of polyploid cells is less.
WT CC
[103]
A
KRP1
CC arrest and inhibition of cell proliferation, G2 phase is
longer; a weak overexpression of KRP2 led to an increment
in DNA ploidy.
Attractor with period 2 oscillating between G1 and G1/S
transition.
[21, 30,
126]
PA
APC/C
Gain-of-function of APC/C subunit CCS52A2 enhanced
endoreduplication entry; more cells with increased DNA
ploidy.
Cyclic attractor pointing to endocycle.
[44]
A
Summary of mutant phenotypes and recovered attractors simulating that mutation. A means that the result of simulation is in Agreement with the data
available in the reference(s). PA means it is Partially Agrees with evidence, because not all expected traits were reproduced by the attractor but this does
not contradict the mutant phenotype. NR are attractors that do not make sense with expected behavior and therefore, the model did Not Recover the
mutant phenotype.
doi:10.1371/journal.pcbi.1004486.t003
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
14 / 28
endoreduplication attractors of CDKB1;1 loss-of-function or E2Fa gain-of-function, where
E2Fb, E2Fc and MYB factors expression states alternate between “ON” and “OFF” (Fig 5).
We highlight APC/C gain-of-function simulations, as it provides a possible mechanism for
plant hormones action over the CC machinery and, thus how such key morphogens regulate
cell proliferation patterns. Recently, Takahashi and collaborators reported a direct connection
between cytokinins and CC machinery in A. thaliana root [131]. The authors showed that
ARR2, a transcriptional factor of cytokinins signaling, induces expression of APC/C activator
protein CCS52A1. Our simulated APC/C gain-of-function is congruent with that observation,
since it reproduces the configuration attained by a cell entering an endocycle when APC/C
activity is enhanced (Fig 5C), as it happens at the elongation zone of A. thaliana root. There-
fore, our model is able to recover the attractors of loss- and gain-of-function mutant pheno-
types reported experimentally, and it thus provides a mechanistic explanation for observed
patterns of expression in both normal CC and during endoreduplication cycles or endocycle.
Table 4. Phenotypes of loss-of-function mutations in CC components.
Node
Phenotypes of loss of function
Recovered attractor(s)
Refs.
Model
CYCD3;1
When this cyclin is depleted by sucrose starvation, cells are
arrested in G1 phase; in adult leaves, triple mutant of
cycd3;1–3 led to a diminished number of cells.
Period 2 oscillating between G1 and G1/S transition.
[23,
92]
A
SCF
Plants with a diminished level of SKP2 do not show obvious
affected development but KRP1 is accumulated.
Similar to a normal CC but endoreduplication would be
favored by the KRP1 stabilization.
[93]
A
RBR
Proliferation is promoted and cell differentiation is impaired;
downregulation of RBR in rice promotes an increase of cells
in S-phase.
One attractor of a normal CC (includes 81.98% of possible
configurations) and other attractor oscillates among
G2-S-G2 (18.02% of configurations).
[127]
A
E2Fa
Expression of E2Fb, RBR and other CC regulators decrease;
more cells in G1 and G2 with respect to WT.
Fixed-point attractor with E2Fe and CYCD3;1 present
suggesting an arrest in a Gap phase.
[90]
PA
E2Fb
Without information.
Fixed-point attractor representing the G1/S transition.
-
-
E2Fc
Mitotic proteins such as CYCB1;1 have increased
expression, ploidy is reduced.
Fixed-point attractor of M phase arrest.
[37,
91]
PA
E2Fe
Increased endoreduplication.
Attractor of endoreduplication (period 7).
[44]
A
MYB77
Lower density of lateral roots, inconclusive data to evaluate
simulation.
CC of seven configurations.
[76]
-
MYB3R1/
4
Lower levels of G2/M transcripts, incomplete cell division,
some embryos only have one cell with multiple nuclei.
2 attractors, the first seems a three-configurations
endocycle, and the second is a CC of seven configurations
where APC/C is always absent.
[43]
A
CYCB1;1
Cyclin widely used as a marker of cell proliferation, its
absence is associated with differentiated cells.
Attractor characterizing endocycle (period 8), intriguingly
APC/C is never present.
[128]
-
CDKB1;1
Overexpression of a dominant negative allele leads to
enhanced endoreduplication.
Attractor of endoreduplication (period 11).
[129]
A
CYCA2;3
In null mutants, cells with 2C DNA content decreases before
than in WT, endocycles begin before and are faster than in
WT.
Attractor which is an endocycle (period 7).
[103]
PA
KRP1
No evident phenotypic effects observed but relative kinase
activity increases to 1.5 in relation to WT.
A CC without alterations.
[114]
A
APC/C
Loss of CCS52A2 function (activator subunit of APC/C)
produces a decrement in the number of meristematic cells
without affecting endoreduplication index; cells in quiescent
center become mitotically active.
Fixed-point attractor of a CC arrest previous to conclude
mitosis.
[109]
PA
Summary of mutant phenotypes and recovered attractor when that mutation was simulated. Abbreviations in Model column are as in Table 3.
doi:10.1371/journal.pcbi.1004486.t004
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
15 / 28
Plant E2Fc and KRP1: validation of A. thaliana CC GRN
We test if the CC GRN recovers the periodic patterns observed in synchronization experiments
of A. thaliana CC molecular components. Interestingly, the E2Fc repressor and KRP1 are regu-
lators that have two short lapses of expression in the attractor recovered in the continuous
model (Fig 6), and experimentally they also show two peaks of expression when synchronized
with aphidicolin [74]. In such synchronization experiments, the expression of E2Fc increases
Fig 5. Attractors recovered by simulations of loss- or gain-of-function mutants of four CC components. (A) The simulation of loss of CDKB1;1
function produced only one cyclic attractor with period 7 that resembles G1 ! S ! G2 ! G1 cycle, whereas in (B) with simulation of loss of KRP1 function,
one cyclic attractor was attained, which has period 11 and comprises 100% of the initial conditions. This attractor is almost identical to WT phenotype but
without KRP1. With the simulation of APC/C gain-of-function, a single attractor with period 7 was recovered, which is shown in (C) and is consistent with an
endoreduplication cycle. Attractors obtained with the simulation of E2Fa overexpression are shown in (D). Two attractors were found, one of them has period
10 and the 40.48% of the initial conditions converge to that cycle that is closely similar to the WT CC attractor. The second attractor that correspond to E2Fa
overexpression has period 8 and it is very similar to the endoreduplication attractor of loss of CDKB1;1 function, which comprises 59.52% of possible network
configurations.
doi:10.1371/journal.pcbi.1004486.g005
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
16 / 28
from late S to middle G2, but then it decreases dramatically in late G2. In the model, E2Fc
appears from S to G2 phase, and then a second increment of E2Fc expression in G2/M is
observed. The latter correspondence is a further validation of the CC GRN model proposed
here. Furthermore, synchronization experiments using sucrose have shown that KRP1 is
expressed previous to G1/S transition and before mitosis [132], in a similar way that occurs in
the model. More recently it has been proposed that KRP1 has a role during G1/S and G2/M
transitions [93]; the latter should be important for endoreduplication control [78]. Once again,
such roles and expression profiles are consistent with the recovered active state of KRP1 in our
model.
In contrast with the consistent behaviors of E2Fc and KRP1 components to recovered
results with our model, E2Fe results do not coincide with previous observations. In our model
this E2F factor presents only one peak from S to early M phase, but according to synchroniza-
tion experiments [69], E2Fe has two peaks of expression. One of its peaks is due to regulation
by other E2F family factors during S phase, while the G2/M peak could be due to MSA ele-
ments. Indeed, when the regulatory motifs for E2F binding are deleted from E2Fe, it can still be
expressed although at lower levels [96], suggesting that additional transcription factors regulate
its expression. Such factors could belong to the MYB family as suggested for the A. thaliana
CC GRN proposed here.
Discussion
The canonical cyclic behavior of eukaryotic cells as they go from DNA duplication to cytokine-
sis suggests that a conserved underlying mechanism with shared molecular components and/
or regulatory logic should exist. While yeast and animal CC have been thoroughly studied and
modelled, plant CC is less studied and no comprehensive model for it has been proposed.
In this study we put forward a Boolean model of the A. thaliana CC GRN. We show that
this model robustly recovers a single cyclic attractor or steady state with 11 network configura-
tions. Such configurations correspond to those observed experimentally for the CC compo-
nents included here at each one of the CC stages. In addition, the canonical order of sequential
transitions that is recovered also mimics the observed temporal pattern of transition from one
Fig 6. Dynamical behavior of E2Fc and KRP1 according to the continuous model. These nodes were chosen by their peculiar pattern of expression,
which was qualitatively recovered by the Boolean and continuous models.
doi:10.1371/journal.pcbi.1004486.g006
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
17 / 28
configuration to another one along the CC (Fig 2). The fact that the 16,384 initial conditions of
the proposed system converge to this single cyclic attractor already suggests that the GRN com-
prises a robust module that integrates the necessary and sufficient set of components and inter-
actions to recover molecular oscillations experimentally observed. The proposed GRN is also
robust to alterations, being similarly robust to previously published models for other cell differ-
entiation or developmental modules [116, 117, 133]. The model is validated because it recovers
A. thaliana wild type and altered (in gain- and loss-of-function) configurations and cycling
behaviors. The comparison between experimentally observed and recovered gene configura-
tions is summarized in Tables 3 and 4.
Some cyclins such as CYCD3;1 and CYCB1;1, important components during G1/S and G2/
M transitions, show a mutually exclusive regulation, as occurs in a predator-prey Lotka-Vol-
terra dynamical system [134], even though they do not interact directly. Their mutual exclu-
sion is achieved thanks to the coordinated expression of genes with specific proteolytic
degradation capacity. Our cyclic attractor shows two transcriptional periods, one of them in S-
phase regulated by E2F-RBR pathway, and the second one operating at a time previous to M-
phase and regulated by MYB transcription factors. The SCF and APC/C ubiquitin ligases work
during G2-to-M phases, and during mitosis exit, respectively. Therefore, the fourteen nodes
and their interactions proposed in the CC GRN constitute a necessary and sufficient set of
restrictions to recover the oscillations of node states characteristic of CC phases.
Two alternative possibilities could drive CC progression in actual organisms. The first
would imply that transitions from one CC state to the next would require external cues, like
the cell size. The alternative possibility is that CC progression and the temporal pattern of tran-
sitions among stages are both emergent consequences of an underlying complex regulatory
network, and do not require external cues, or these only reinforce such temporal progression
emergent from complex underlying regulatory interactions. Our CC GRN model supports the
latter. This does not imply that several internal or external signals or molecules, such as hor-
mones or other types of cues could alter the CC. Therefore, the two alternative possibilities are
not exclusive but they likely complement or enhance each other. Indeed, A. thaliana CC is reg-
ulated by plant hormones, light, sucrose, osmotic stress [135] or oxidative stress [136]. These
could now be modelled as CC modulators.
In the model proposed here we avoided redundancy. For instance, the KRP1 node repre-
sents the KRP family members that share several functions. Also the metaphase-anaphase tran-
sition could be added to the model when more data about APC/C regulation (i.e. negative
feedback loop comprising CDK-cyclin complexes, or the regulation of Cdc20 homologues)
becomes available in plants. Apparently, these simplifications did not disrupt the main features
of the A. thaliana CC, since the cyclic behavior distinctive of the CC components was correctly
recovered.
A mechanistic model for the A. thaliana CC: novel predictions
Our proposed GRN model suggests some predictions regarding the regulation of certain CC
components in A. thaliana. Such predictions can be classified into two types. The first type per-
tains to those recovered by in silico promoter analysis. The predictions of the second type were
inferred from data of other eukaryotes, because they seem to imply conserved components and
some evidence from A. thaliana suggested that these interactions are part of the CC GRN in A.
thaliana. Three interactions belong to the second type, E2Fb ! SCF, CDKB1;1-CYCA2;3 a
E2Fa and APC/C a SCF (see Table 1 for a synthesis of hypothetical interactions). Although
some evidence supports the idea that these interactions could exist in A. thaliana, they should
be corroborated with additional experimental examination.
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
18 / 28
Our model provides a dynamic explanation to the cyclic behavior of certain transcription
factors and predicts a novel interaction for E2F and MYB regulators; they connect waves of
periodic expression that seem to be key for the robust limit cycle attractor that characterizes
CC behavior. Interestingly, previous studies have shown that such periodic transcription can
be maintained even in the absence of S-phase and mitotic cyclins [4], which underpin the role
of a transcription factor network oscillator for the correct CC progression [137]. A regulatory
interaction between E2F and MYB factors (or among the equivalent regulators) may be con-
served among other eukaryotes (e.g. mammals and yeast), but there is no experimental support
yet for it in A. thaliana. After looking for the same direct evidence in A. thaliana and not find-
ing it, we thought about an alternative regulatory mechanism that consists in transcription fac-
tors acting between E2F and MYB. Hence, we decided to analyze the important transcription
factor families known so far, to find out if one of their members could be mediating the regula-
tion between E2F and MYB. The TCP (for Teosinte branched 1, Cycloidea, PCF) and the MYB
family were chosen because they have been reported to be involved in CC regulation [42].
Based on their gene expression patterns and promoter sequence analysis, MYB77 was our best
candidate: it is expressed at the beginning of M phase, and could be regulated by E2F and regu-
lator of MYB (see Table 1). A second possibility might be that several tissue-specific transcrip-
tion factors are involved in E2F-MYB genetic regulation (e.g. GL3, MYB88, SHR/SCR [17],
MYB59 [138] or even members of the MADS box gene family could be implied). Indeed, we
have recently documented that a MADS-box gene, XAL1, encodes a transcription factor that
regulates several CC components (García-Cruz et al., in preparation).
A. thaliana CC in comparison to animal and yeast CC
Differences among eukaryotic CCs allow us to recognize or characterize alternative mecha-
nisms for the regulation of CC. The first difference between GRN of A. thaliana CC and that of
other eukaryotes, concerns the number of duplicates of some key regulators. A. thaliana has up
to ten copies of some of the genes that encode for CC regulators (e.g. families of cyclins or
CDK), while yeast, mammals or the algae Ostreococcus tauri, have much fewer duplicates [20,
139–141]. The only exception concerns the homologues of Retinoblastoma protein, of which
there are three members in humans and mouse, and only one copy in A. thaliana [127]. Future
models should address the explicit role of CC duplicated components in the plastic response of
plant development to environmental conditions. Being sessile, such developmental adjust-
ments, as plants grow under varying environments, are expected to be more important, com-
plex and dynamic than in motile yeast and animals. One possibility is that different members
of the same gene family are linked to different transduction pathways of signals that modulate
CC dynamics.
The second difference among A. thaliana and other CC was regarding the transcriptional
regulation throughout the GRN underlying it. For instance, S. cerevisiae does not have RBR or
E2F homologues, but instead has Whi5, Swi4,6 and Mbp1 proteins which perform equivalent
regulatory functions to the former CC components [142, 143]. S. cerevisiae does not have any
MYB transcription factors but it presents other transcriptional regulators, such as Fkh1/2,
Ndd1 and Mcm1 [142, 144, 145], which regulate the G2/M transition in a similar way to MYBs
in mammals.
Contrary to the conservation in G1/S transition [15, 67], molecular components controlling
G2/M transition seem to vary among different eukaryotes. It seems that molecules such as
WEE1 kinase and CDC25 phosphatase are not conserved. In A. thaliana, CDC25-like has
phosphatase and arsenate-reductase functions [146], while A. thaliana WEE1 phosphorylates
monomeric CDKA;1 in vitro [147], and Nicotiana tabacum WEE1 inhibits CDK activity in
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
19 / 28
vitro [148]. However the lack of any obvious mutant phenotype of CDC25 or WEE1 loss-of-
function mutants predicts that these genes are not involved in the regulation of a normal CC.
Additionally, although WEE1 has a role during DNA damage [146, 149], does not seem to
have a CDKA;1 recognition domain [150]. CDC25-like does not have the required sites for
CDKA;1 recognition [150]. In summary, the positive regulatory feedback between CDKA;1
and CDC25-like, as well as the mutual-inhibitory feedback loop between CDKA;1 and WEE1,
seem not to be conserved in A. thaliana.
Given all that evidence for G2/M regulation, we integrated the regulatory interactions
between stoichiometric CDK inhibitor (KRP1), B-type plant specific CDK and MYB transcrip-
tional factors. It is not surprising that there are clear differences between plant G2 phase regula-
tion and that of other organisms, because variations in this control point could define cell fate.
Although differences among the A. thaliana CC GRN uncovered here and that of yeasts and
animals have now become clear, we think that the basic regulatory CC module reported here,
will be a useful framework to incorporate and discover new components of the CC GRNs in
plants and also in other eukaryotes.
Despite the fact that our CC GRN model recovers observed CC stage configurations and
their canonical pattern of temporal transitions, it did not recover an alternative attractor that
corresponds to the endocycle. We hypothesize that the same multi-stable GRN underlies both
states, and additional components yet to be connected to the CC GRN will ensure a cyclic
attractor corresponding to the complete CC, and another one with shorter period correspond-
ing to the endocycle. In its present form, our model suggests that CYCD3;1 function, which
has been associated with the proliferative state [108] and with a delay in the endocycle onset
[23], is important to enter the endocycle. Besides, it also has been reported that CYCD3;1 plays
a role in G1/S transition [121] and regulates RBR protein during DNA replication [89]. Fur-
thermore, the endoreduplication attractor obtained in some of our mutant simulations (e.g. Fig
5A, 5C and 5D-right) also supports the role of CYCD3;1 in entering an endocycle.
The GRN model of A. thaliana CC could help to identify physiological or developmental
interactions involved in the tight relationship between proliferation and differentiation observed
during different stages of development [1, 88, 108, 109, 126]. Previous to cell division, the cell
senses its intracellular and environmental conditions to arrest or promote CC progress. Such
cues directly affect the CC machinery, which does not depend on a master or central regulator.
CC control is the result of a network formed by feedback and feedforward loops between
complexes of CDK-cyclin and its regulators. It is not evident how complex dynamical processes
such as CC progression emerge from simple interactions among components acting simulta-
neously. The proposed CC GRN will be very helpful to study how cell proliferation/differentia-
tion decisions and balance keeps a suitable spatio-temporal control of CC during plant growth
and development.
Supporting Information
S1 Text. Logical rules of A. thaliana CC Boolean model.
(PDF)
S2 Text. Equations, parameters, analysis of parameters and initial conditions of the contin-
uous version of A. thaliana CC model.
(PDF)
S3 Text. New recovered attractors by robustness analysis. Additional attractors yielded by
making alterations in each bit of logical functions.
(PDF)
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
20 / 28
S4 Text. Attractors obtained in the simulation of mutant phenotypes.
(PDF)
Acknowledgments
The present manuscript is part of EOG’s PhD thesis in the Graduate Program in Biomedical
Sciences of the Universidad Nacional Autónoma de México (UNAM). EOG acknowledges the
scholarship and financial support provided by Consejo Nacional de Ciencia y Tecnología of
Mexico (CONACyT). This work greatly benefited from input provided by Dr. Joseph G.
Dubrovsky. We also thank Elizabeth Gilbert for editing previous versions of the paper; remain-
ing errors are our responsability. We acknowledge the help from Diana Romo with various
logistical and technical tasks.
Author Contributions
Analyzed the data: AC EA EOG ERAB MPS KGC. Wrote the paper: EOG ERAB MPS. Con-
ceived and coordinated the study and established the overall logic and core questions to be
addressed: ERAB Conceived and planned the modeling approaches and specific analyses to be
done: EOG ERAB Recovered all the information from the literature: AC EA EOG MPS KGC
Established the logical functions: EA EOG Programmed and ran all the modeling and analyses:
EA EOG.
References
1.
Ramirez-Parra E, Desvoyes B, Gutierrez C. Balance between cell division and differentiation during
plant development. Int J Dev Biol. 2005; 49:467–77. doi: 10.1387/ijdb.052001er PMID: 16096957
2.
Dudits D, Abrahám E, Miskolczi P, Ayaydin F, Bilgin M, Horváth GV. Cell-cycle control as a target for
calcium, hormonal and developmental signals: the role of phosphorylation in the retinoblastoma-cen-
tred pathway. Ann Bot. 2011 5; 107(7):1193–202. doi: 10.1093/aob/mcr038 PMID: 21441245
3.
Ubeda-Tomas S, Beemster GT, Bennett MJ. Hormonal regulation of root growth: integrating local
activities into global behaviour. Trends Plant Sci. 2012;.
4.
Orlando DA, Lin CY, Bernard A, Wang JY, Socolar JES, Iversen ES, et al. Global control of cell-cycle
transcription by coupled CDK and network oscillators. Nature. 2008 6; 453(7197):944–7. doi: 10.
1038/nature06955 PMID: 18463633
5.
Azpeitia E, Benítez M, Padilla-Longoria P, Espinosa-Soto C, Alvarez-Buylla ER. Dynamic network-
based epistasis analysis: Boolean examples. Front Plant Sci. 2011; 2(92). doi: 10.3389/fpls.2011.
00092 PMID: 22645556
6.
Wang RS, Saadatpour A, Albert R. Boolean modeling in systems biology: an overview of methodology
and applications. Phys Biol. 2012 10; 9(5):055001. doi: 10.1088/1478-3975/9/5/055001 PMID:
23011283
7.
Tyson JJ, Novak B. Regulation of the eukaryotic cell cycle: molecular antagonism, hysteresis, and
irreversible transitions. J Theor Biol. 2001; 210(2):249–63. doi: 10.1006/jtbi.2001.2293 PMID:
11371178
8.
Guardavaccaro D, Pagano M. Stabilizers and destabilizers controlling cell cycle oscillators. Mol Cell.
2006; 22(1):1–4. doi: 10.1016/j.molcel.2006.03.017 PMID: 16600864
9.
Zhang T, Schmierer B, Novak B. Cell cycle commitment in budding yeast emerges from the coopera-
tion of multiple bistable switches. Open Biol. 2011; 1(3). doi: 10.1098/rsob.110009 PMID: 22645649
10.
Kapuy O, He E, Lopez-Aviles S, Uhlmann F, Tyson JJ, Novak B. System-level feedbacks control cell
cycle progression. FEBS Lett. 2009; 583(24):3992–8. doi: 10.1016/j.febslet.2009.08.023 PMID:
19703449
11.
Ingolia N. Cell cycle: bistability is needed for robust cycling. Curr Biol. 2005 12; 15(23):R961–3. doi:
10.1016/j.cub.2005.11.012 PMID: 16332526
12.
Pomerening JR, Kim SY, Ferrell JE. Systems-level dissection of the cell-cycle oscillator: bypassing
positive feedback produces damped oscillations. Cell. 2005 8; 122(4):565–78. doi: 10.1016/j.cell.
2005.06.016 PMID: 16122424
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
21 / 28
13.
Charvin G, Oikonomou C, Siggia ED, Cross FR. Origin of irreversibility of cell cycle start in budding
yeast. PLoS Biol. 2010 1; 8(1):e1000284. doi: 10.1371/journal.pbio.1000284 PMID: 20087409
14.
Yao G, Tan C, West M, Nevins JR, You L. Origin of bistability underlying mammalian cell cycle entry.
Mol Syst Biol. 2011; 7:485. doi: 10.1038/msb.2011.19 PMID: 21525871
15.
Cross FR, Buchler NE, Skotheim JM. Evolution of networks and sequences in eukaryotic cell cycle
control. Philos Trans R Soc Lond B Biol Sci. 2011; 366(1584):3532–44. doi: 10.1098/rstb.2011.0078
PMID: 22084380
16.
Jensen LJ, Jensen TS, de Lichtenberg U, Brunak S, Bork P. Co-evolution of transcriptional and post-
translational cell-cycle regulation. Nature. 2006 10; 443(7111):594–7. PMID: 17006448
17.
Komaki S, Sugimoto K. Control of the plant cell cycle by developmental and environmental cues.
Plant Cell Physiol. 2012 6; 53(6):953–64. doi: 10.1093/pcp/pcs070 PMID: 22555815
18.
Peeper DS, Parker LL, Ewen ME, Toebes M, Hall FL, Xu M, et al. A- and B-type cyclins differentially
modulate substrate specificity of cyclin-cdk complexes. EMBO J. 1993 5; 12(5):1947–54. PMID:
8491188
19.
Nieuwland J, Scofield S, Murray JAH. Control of division and differentiation of plant stem cells and
their derivatives. Semin Cell Dev Biol. 2009 12; 20(9):1134–42. doi: 10.1016/j.semcdb.2009.09.011
PMID: 19770062
20.
Inzé D, De Veylder L. Cell cycle regulation in plant development. Annu Rev Genet. 2006; 40:77–105.
doi: 10.1146/annurev.genet.40.110405.090431 PMID: 17094738
21.
Verkest A, Manes CL, Vercruysse S, Maes S, Van Der Schueren E, Beeckman T, et al. The cyclin-
dependent kinase inhibitor KRP2 controls the onset of the endoreduplication cycle during Arabidopsis
leaf development through inhibition of mitotic CDKA;1 kinase complexes. Plant Cell. 2005; 17
(6):1723–36. doi: 10.1105/tpc.105.032383 PMID: 15863515
22.
Beemster GT, De Veylder L, Vercruysse S, West G, Rombaut D, Van Hummelen P, et al. Genome-
wide analysis of gene expression profiles associated with cell cycle transitions in growing organs of
Arabidopsis. Plant Physiol. 2005 6; 138(2):734–43. doi: 10.1104/pp.104.053884 PMID: 15863702
23.
Dewitte W, Scofield S, Alcasabas AA, Maughan SC, Menges M, Braun N, et al. Arabidopsis CYCD3
D-type cyclins link cell proliferation and endocycles and are rate-limiting for cytokinin responses. Proc
Natl Acad Sci U S A. 2007; 104(36):14537–42. doi: 10.1073/pnas.0704166104 PMID: 17726100
24.
Boruc J, Inze D, Russinova E. A high-throughput bimolecular fluorescence complementation protein-
protein interaction screen identifies functional Arabidopsis CDKA/B-CYCD4/5 complexes. Plant Sig-
nal Behav. 2010; 5(10):1276–81. doi: 10.4161/psb.5.10.13037 PMID: 20861687
25.
Van Leene J, Hollunder J, Eeckhout D, Persiau G, Van De Slijke E, Stals H, et al. Targeted interac-
tomics reveals a complex core cell cycle machinery in Arabidopsis thaliana. Mol Syst Biol. 2010;
6:397. doi: 10.1038/msb.2010.53 PMID: 20706207
26.
Weingartner M, Criqui MC, Meszaros T, Binarova P, Schmit AC, Helfer A, et al. Expression of a non-
degradable cyclin B1 affects plant development and leads to endomitosis by inhibiting the formation
of a phragmoplast. Plant Cell. 2004; 16(3):643–57. doi: 10.1105/tpc.020057 PMID: 15004270
27.
Boudolf V, Lammens T, Boruc J, Van Leene J, Van Den Daele H, Maes S, et al. CDKB1;1 forms a
functional complex with CYCA2;3 to suppress endocycle onset. Plant Physiol. 2009; 150(3):1482–93.
doi: 10.1104/pp.109.140269 PMID: 19458112
28.
Vanneste S, Coppens F, Lee E, Donner TJ, Xie Z, Isterdael GV, et al. Developmental regulation of
CYCA2s contributes to tissue-specific proliferation in Arabidopsis. EMBO J. 2011 8; 30(16):3430–41.
doi: 10.1038/emboj.2011.240 PMID: 21772250
29.
De Veylder L, Beeckman T, Beemster GT, Krols L, Terras F, Landrieu I, et al. Functional analysis of
cyclin-dependent kinase inhibitors of Arabidopsis. Plant Cell. 2001 7; 13(7):1653–68. doi: 10.1105/
TPC.010087 PMID: 11449057
30.
Wang H, Zhou Y, Bird DA, Fowke LC. Functions, regulation and cellular localization of plant cyclin-
dependent kinase inhibitors. J Microsc. 2008 8; 231(2):234–46. doi: 10.1111/j.1365-2818.2008.
02039.x PMID: 18778421
31.
Verkest A, Weinl C, Inze D, De Veylder L, Schnittger A. Switching the cell cycle. Kip-related proteins
in plant cell cycle control. Plant Physiol. 2005; 139(3):1099–106. doi: 10.1104/pp.105.069906 PMID:
16286449
32.
De Veylder L, Joubes J, Inze D. Plant cell cycle transitions. Curr Opin Plant Biol. 2003; 6(6):536–43.
doi: 10.1016/j.pbi.2003.09.001 PMID: 14611951
33.
Gutierrez C, Ramirez-Parra E, Castellano MM, del Pozo JC. G1 to S transition: more than a cell cycle
engine switch. Curr Opin Plant Biol. 2002; 5(6):480–6. doi: 10.1016/S1369-5266(02)00301-1 PMID:
12393009
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
22 / 28
34.
de Jager SM, Menges M, Bauer UM, Murra JA. Arabidopsis E2F1 binds a sequence present in the
promoter of S-phase-regulated gene AtCDC6 and is a member of a multigene family with differential
activities. Plant Mol Biol. 2001 11; 47(4):555–68. doi: 10.1023/A:1011848528377 PMID: 11669580
35.
del Pozo JC, Boniotti MB, Gutierrez C. Arabidopsis E2Fc functions in cell division and is degraded by
the ubiquitin-SCF(AtSKP2) pathway in response to light. Plant Cell. 2002; 14(12):3057–71. doi: 10.
1105/tpc.006791 PMID: 12468727
36.
Henley SA, Dick FA. The retinoblastoma family of proteins and their regulatory functions in the mam-
malian cell division cycle. Cell Div. 2012; 7(1):10. doi: 10.1186/1747-1028-7-10 PMID: 22417103
37.
del Pozo JC, Diaz-Trivino S, Cisneros N, Gutierrez C. The balance between cell division and endore-
plication depends on E2FC-DPB, transcription factors regulated by the ubiquitin-SCFSKP2A pathway
in Arabidopsis. Plant Cell. 2006; 18(9):2224–35. doi: 10.1105/tpc.105.039651 PMID: 16920782
38.
Planchais S, Perennes C, Glab N, Mironov V, Inze D, Bergounioux C. Characterization of cis-acting
element involved in cell cycle phase-independent activation of Arath;CycB1;1 transcription and identi-
fication of putative regulatory proteins. Plant Mol Biol. 2002; 50(1):111–27. doi: 10.1023/
A:1016018711532 PMID: 12139003
39.
Zhu W, Giangrande PH, Nevins JR. E2Fs link the control of G1/S and G2/M transcription. EMBO J.
2004 11; 23(23):4615–26. doi: 10.1038/sj.emboj.7600459 PMID: 15510213
40.
Ito M. Conservation and diversification of three-repeat Myb transcription factors in plants. J Plant Res.
2005; 118(1):61–9. doi: 10.1007/s10265-005-0192-8 PMID: 15703854
41.
Nakata Y, Shetzline S, Sakashita C, Kalota A, Rallapalli R, Rudnick SI, et al. c-Myb contributes to G2/
M cell cycle transition in human hematopoietic cells by direct regulation of cyclin B1 expression. Mol
Cell Biol. 2007; 27(6):2048–58. doi: 10.1128/MCB.01100-06 PMID: 17242210
42.
Berckmans B, De Veylder L. Transcriptional control of the cell cycle. Curr Opin Plant Biol. 2009 10; 12
(5):599–605. doi: 10.1016/j.pbi.2009.07.005 PMID: 19700366
43.
Haga N, Kobayashi K, Suzuki T, Maeo K, Kubo M, Ohtani M, et al. Mutations in MYB3R1 and
MYB3R4 cause pleiotropic developmental defects and preferential down-regulation of multiple G2/M-
specific genes in Arabidopsis. Plant Physiol. 2011 10; 157(2):706–17. doi: 10.1104/pp.111.180836
PMID: 21862669
44.
Lammens T, Boudolf V, Kheibarshekan L, Zalmas LP, Gaamouche T, Maes S, et al. Atypical E2F
activity restrains APC/CCCS52A2 function obligatory for endocycle onset. Proc Natl Acad Sci U S A.
2008 9; 105(38):14721–6. doi: 10.1073/pnas.0806510105 PMID: 18787127
45.
Chen KC, Calzone L, Csikasz-Nagy A, Cross FR, Novak B, Tyson JJ. Integrative analysis of cell cycle
control in budding yeast. Mol Biol Cell. 2004 8; 15(8):3841–62. doi: 10.1091/mbc.E03-11-0794 PMID:
15169868
46.
Fauré A, Naldi A, Chaouiya C, Thieffry D. Dynamical analysis of a generic Boolean model for the con-
trol of the mammalian cell cycle. Bioinformatics. 2006; 22(14):e124–31. doi: 10.1093/bioinformatics/
btl210 PMID: 16873462
47.
Fauré A, Naldi A, Lopez F, Chaouiya C, Ciliberto A, Thieffry D. Modular logical modelling of the bud-
ding yeast cell cycle. Mol Biosyst. 2009 12; 5(12):1787–96. doi: 10.1039/b910101m PMID: 19763337
48.
Irons DJ. Logical analysis of the budding yeast cell cycle. J Theor Biol. 2009 4; 257(4):543–59. doi:
10.1016/j.jtbi.2008.12.028 PMID: 19185585
49.
Goldbeter A. A minimal cascade model for the mitotic oscillator involving cyclin and cdc2 kinase. Proc
Natl Acad Sci U S A. 1991 10; 88(20):9107–11. doi: 10.1073/pnas.88.20.9107 PMID: 1833774
50.
Novak B, Tyson JJ. Modeling the control of DNA replication in fission yeast. Proc Natl Acad Sci U S A.
1997 8; 94(17):9147–52. doi: 10.1073/pnas.94.17.9147 PMID: 9256450
51.
Novak B, Csikasz-Nagy A, Gyorffy B, Chen K, Tyson JJ. Mathematical model of the fission yeast cell
cycle with checkpoint controls at the G1/S, G2/M and metaphase/anaphase transitions. Biophys
Chem. 1998; 72:185–200. doi: 10.1016/S0301-4622(98)00133-1 PMID: 9652094
52.
Novak B, Csikasz-Nagy A, Gyorffy B, Nasmyth K, Tyson JJ. Model scenarios for evolution of the
eukaryotic cell cycle. Philos Trans R Soc Lond B Biol Sci. 1998; 353(1378):2063–76. doi: 10.1098/
rstb.1998.0352 PMID: 10098216
53.
Novak B, Pataki Z, Ciliberto A, Tyson JJ. Mathematical model of the cell division cycle of fission yeast.
Chaos. 2001; 11(1):277–286. doi: 10.1063/1.1345725 PMID: 12779461
54.
Tyson JJ, Csikasz-Nagy A, Novak B. The dynamics of cell cycle regulation. Bioessays. 2002; 24
(12):1095–109. doi: 10.1002/bies.10191 PMID: 12447975
55.
Li B, Shao B, Yu C, Ouyang Q, Wang H. A mathematical model for cell size control in fission yeast. J
Theor Biol. 2010 6; 264(3):771–81. doi: 10.1016/j.jtbi.2010.03.023 PMID: 20303984
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
23 / 28
56.
Qu Z, MacLellan WR, Weiss JN. Dynamics of the cell cycle: checkpoints, sizers, and timers. Biophys
J. 2003 12; 85(6):3600–11. doi: 10.1016/S0006-3495(03)74778-X PMID: 14645053
57.
Csikász-Nagy A, Battogtokh D, Chen KC, Novák B, Tyson JJ. Analysis of a generic model of eukary-
otic cell-cycle regulation. Biophys J. 2006 6; 90(12):4361–79. doi: 10.1529/biophysj.106.081240
PMID: 16581849
58.
Li F, Long T, Lu Y, Ouyang Q, Tang C. The yeast cell-cycle network is robustly designed. Proc Natl
Acad Sci U S A. 2004; 101(14):4781–6. doi: 10.1073/pnas.0305937101 PMID: 15037758
59.
Davidich MI, Bornholdt S. Boolean network model predicts cell cycle sequence of fission yeast. PLoS
One. 2008; 3(2):e1672. doi: 10.1371/journal.pone.0001672 PMID: 18301750
60.
Hong C, Lee M, Kim D, Kim D, Cho KH, Shin I. A checkpoints capturing timing-robust Boolean model
of the budding yeast cell cycle regulatory network. BMC Syst Biol. 2012; 6:129. doi: 10.1186/1752-
0509-6-129 PMID: 23017186
61.
Davidich MI, Bornholdt S. Boolean network model predicts knockout mutant phenotypes of fission
yeast. PLoS One. 2013; 8(9):e71786. doi: 10.1371/journal.pone.0071786 PMID: 24069138
62.
Singhania R, Sramkoski RM, Jacobberger JW, Tyson JJ. A hybrid model of mammalian cell cycle reg-
ulation. PLoS Comput Biol. 2011; 7(2):e1001077. doi: 10.1371/journal.pcbi.1001077 PMID:
21347318
63.
Srividhya J, Gopinathan MS. A simple time delay model for eukaryotic cell cycle. J Theor Biol. 2006 8;
241(3):617–27. doi: 10.1016/j.jtbi.2005.12.020 PMID: 16473373
64.
Li C, Wang J. Landscape and flux reveal a new global view and physical quantification of mammalian
cell cycle. Proc Natl Acad Sci U S A. 2014 9; 111(39):14130–5. doi: 10.1073/pnas.1408628111
PMID: 25228772
65.
Li C, Wang J. Quantifying the underlying landscape and paths of cancer. J R Soc Interface. 2014 11;
11(100):20140774. doi: 10.1098/rsif.2014.0774 PMID: 25232051
66.
Fauré A, Thieffry D. Logical modelling of cell cycle control in eukaryotes: a comparative study. Mol
Biosyst. 2009 12; 5(12):1569–81. doi: 10.1039/b907562n PMID: 19763341
67.
Zhao X, Harashima H, Dissmeyer N, Pusch S, Weimer AK, Bramsiepe J, et al. A General G1/S-Phase
Cell-Cycle Control Module in the Flowering Plant Arabidopsis thaliana. PLoS Genetics. 2012 8; 8(8):
e1002847. doi: 10.1371/journal.pgen.1002847 PMID: 22879821
68.
Adachi S, Nobusawa T, Umeda M. Quantitative and cell type-specific transcriptional regulation of A-
type cyclin-dependent kinase in Arabidopsis thaliana. Dev Biol. 2009; 329(2):306–14. doi: 10.1016/j.
ydbio.2009.03.002 PMID: 19285489
69.
Mariconti L, Pellegrini B, Cantoni R, Stevens R, Bergounioux C, Cella R, et al. The E2F family of tran-
scription factors from Arabidopsis thaliana. Novel and conserved components of the retinoblastoma/
E2F pathway in plants. J Biol Chem. 2002 3; 277(12):9911–9. doi: 10.1074/jbc.M110616200 PMID:
11786543
70.
Zhang L, Wang C. F-box protein Skp2: a novel transcriptional target of E2F. Oncogene. 2006 4; 25
(18):2615–27. doi: 10.1038/sj.onc.1209286 PMID: 16331253
71.
Rustici G, Mata J, Kivinen K, Lio P, Penkett CJ, Burns G, et al. Periodic gene expression program of
the fission yeast cell cycle. Nat Genet. 2004; 36(8):809–17. doi: 10.1038/ng1377 PMID: 15195092
72.
Menges M, Hennig L, Gruissem W, Murray JAH. Cell cycle-regulated gene expression in Arabidopsis.
J Biol Chem. 2002 11; 277(44):41987–2002. doi: 10.1074/jbc.M207570200 PMID: 12169696
73.
Menges M, Hennig L, Gruissem W, Murray JAH. Genome-wide gene expression in an Arabidopsis
cell suspension. Plant Mol Biol. 2003 11; 53(4):423–42. doi: 10.1023/B:PLAN.0000019059.56489.ca
PMID: 15010610
74.
Menges M, de Jager SM, Gruissem W, Murray JA. Global analysis of the core cell cycle regulators of
Arabidopsis identifies novel genes, reveals multiple and highly specific profiles of expression and pro-
vides a coherent model for plant cell cycle control. Plant J. 2005; 41(4):546–66. doi: 10.1111/j.1365-
313X.2004.02319.x PMID: 15686519
75.
Romero I, Fuertes A, Benito MJ, Malpica JM, Leyva A, Paz-Ares J. More than 80R2R3-MYB regula-
tory genes in the genome of Arabidopsis thaliana. Plant J. 1998 5; 14(3):273–84. doi: 10.1046/j.1365-
313X.1998.00113.x PMID: 9628022
76.
Shin R, Burch AY, Huppert KA, Tiwari SB, Murphy AS, Guilfoyle TJ, et al. The Arabidopsis transcrip-
tion factor MYB77 modulates auxin signal transduction. Plant Cell. 2007 8; 19(8):2440–53. doi: 10.
1105/tpc.107.050963 PMID: 17675404
77.
Prouse MB, Campbell MM. The interaction between MYB proteins and their target DNA binding sites.
Biochim Biophys Acta. 2012 1; 1819(1):67–77. doi: 10.1016/j.bbagrm.2011.10.010 PMID: 22067744
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
24 / 28
78.
Weinl C, Marquardt S, Kuijt SJH, Nowack MK, Jakoby MJ, Hülskamp M, et al. Novel functions of plant
cyclin-dependent kinase inhibitors, ICK1/KRP1, can act non-cell-autonomously and inhibit entry into
mitosis. Plant Cell. 2005 6; 17(6):1704–22. doi: 10.1105/tpc.104.030486 PMID: 15749764
79.
Magyar Z, De Veylder L, Atanassova A, Bakó L, Inzé D, Bögre L. The role of the Arabidopsis E2FB
transcription factor in regulating auxin-dependent cell division. Plant Cell. 2005 9; 17(9):2527–41. doi:
10.1105/tpc.105.033761 PMID: 16055635
80.
Sozzani R, Maggio C, Varotto S, Canova S, Bergounioux C, Albani D, et al. Interplay between Arabi-
dopsis activating factors E2Fb and E2Fa in cell cycle progression and development. Plant Physiol.
2006 4; 140(4):1355–66. doi: 10.1104/pp.106.077990 PMID: 16514015
81.
Leone G, DeGregori J, Yan Z, Jakoi L, Ishida S, Williams RS, et al. E2F3 activity is regulated during
the cell cycle and is required for the induction of S phase. Genes Dev. 1998 7; 12(14):2120–30. doi:
10.1101/gad.12.14.2120 PMID: 9679057
82.
Peters JM. SCF and APC: the Yin and Yang of cell cycle regulated proteolysis. Curr Opin Cell Biol.
1998; 10(6):759–68. doi: 10.1016/S0955-0674(98)80119-1 PMID: 9914180
83.
Vodermaier HC. APC/C and SCF: controlling each other and the cell cycle. Curr Biol. 2004 9; 14(18):
R787–96. doi: 10.1016/j.cub.2004.09.020 PMID: 15380093
84.
Bashir T, Dorrello NV, Amador V, Guardavaccaro D, Pagano M. Control of the SCF(Skp2-Cks1) ubi-
quitin ligase by the APC/C(Cdh1) ubiquitin ligase. Nature. 2004 3; 428(6979):190–3. doi: 10.1038/
nature02330 PMID: 15014502
85.
Marrocco K, Bergdoll M, Achard P, Criqui MC, Genschik P. Selective proteolysis sets the tempo of the
cell cycle. Curr Opin Plant Biol. 2010; 13(6):631–9. doi: 10.1016/j.pbi.2010.07.004 PMID: 20810305
86.
Boniotti MB, Gutierrez C. A cell-cycle-regulated kinase activity phosphorylates plant retinoblastoma
protein and contains, in Arabidopsis, a CDKA/cyclin D complex. Plant J. 2001 11; 28(3):341–50. doi:
10.1046/j.1365-313X.2001.01160.x PMID: 11722776
87.
Dewitte W, Murray JA. The plant cell cycle. Annu Rev Plant Biol. 2003; 54:235–64. doi: 10.1146/
annurev.arplant.54.031902.134836 PMID: 14502991
88.
Wildwater M, Campilho A, Perez-Perez JM, Heidstra R, Blilou I, Korthout H, et al. The RETINOBLAS-
TOMA-RELATED gene regulates stem cell maintenance in Arabidopsis roots. Cell. 2005 12; 123
(7):1337–49. doi: 10.1016/j.cell.2005.09.042 PMID: 16377572
89.
Nowack MK, Harashima H, Dissmeyer N, Zhao X, Bouyer D, Weimer AK, et al. Genetic framework of
cyclin-dependent kinase function in Arabidopsis. Dev Cell. 2012 5; 22(5):1030–40. doi: 10.1016/j.
devcel.2012.02.015 PMID: 22595674
90.
Magyar Z, Horvath B, Khan S, Mohammed B, Henriques R, De Veylder L, et al. Arabidopsis E2FA
stimulates proliferation and endocycle separately through RBR-bound and RBR-free complexes.
EMBO J. 2012; 31(6):1480–93. doi: 10.1038/emboj.2012.13 PMID: 22307083
91.
Del Pozo JC, Diaz-Trivino S, Cisneros N, Gutierrez C. The E2FC-DPB Transcription Factor Controls
Cell Division, Endoreplication and Lateral Root Formation in a SCF-Dependent Manner. Plant Signal
Behav. 2007; 2(4):273–4. doi: 10.4161/psb.2.4.3897 PMID: 19704635
92.
Planchais S, Samland AK, Murray JA. Differential stability of Arabidopsis D-type cyclins: CYCD3;1 is
a highly unstable protein degraded by a proteasome-dependent mechanism. Plant J. 2004; 38
(4):616–25. doi: 10.1111/j.0960-7412.2004.02071.x PMID: 15125768
93.
Ren H, Santner A, del Pozo JC, Murray JA, Estelle M. Degradation of the cyclin-dependent kinase
inhibitor KRP1 is regulated by two different ubiquitin E3 ligases. Plant J. 2008; 53(5):705–16. doi: 10.
1111/j.1365-313X.2007.03370.x PMID: 18005227
94.
Vandepoele K, Vlieghe K, Florquin K, Hennig L, Beemster GT, Gruissem W, et al. Genome-wide iden-
tification of potential plant E2F target genes. Plant Physiol. 2005; 139(1):316–28. doi: 10.1104/pp.
105.066290 PMID: 16126853
95.
He SS, Liu J, Xie Z, O’Neill D, Dotson S. Arabidopsis E2Fa plays a bimodal role in regulating cell divi-
sion and cell growth. Plant Mol Biol. 2004; 56(2):171–84. doi: 10.1007/s11103-004-2748-8 PMID:
15604736
96.
Berckmans B, Lammens T, Van Den Daele H, Magyar Z, Bögre L, De Veylder L. Light-dependent reg-
ulation of DEL1 is determined by the antagonistic action of E2Fb and E2Fc. Plant Physiol. 2011 11;
157(3):1440–51. doi: 10.1104/pp.111.183384 PMID: 21908689
97.
López-Juez E, Dillon E, Magyar Z, Khan S, Hazeldine S, de Jager SM, et al. Distinct light-initiated
gene expression and cell cycle programs in the shoot apex and cotyledons of Arabidopsis. Plant Cell.
2008 4; 20(4):947–68. doi: 10.1105/tpc.107.057075 PMID: 18424613
98.
Haga N, Kato K, Murase M, Araki S, Kubo M, Demura T, et al. R1R2R3-Myb proteins positively regu-
late cytokinesis through activation of KNOLLE transcription in Arabidopsis thaliana. Development.
2007 3; 134(6):1101–10. doi: 10.1242/dev.02801 PMID: 17287251
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
25 / 28
99.
Araki S, Ito M, Soyano T, Nishihama R, Machida Y. Mitotic cyclins stimulate the activity of c-Myb-like
factors for transactivation of G2/M phase-specific genes in tobacco. J Biol Chem. 2004 7; 279
(31):32979–88. doi: 10.1074/jbc.M403171200 PMID: 15175336
100.
Wang H, Fowke LC, Crosby WL. A plant cyclin-dependent kinase inhibitor gene. Nature. 1997 4; 386
(6624):451–2. doi: 10.1038/386451a0 PMID: 9087400
101.
Wang H, Qi Q, Schorr P, Cutler AJ, Crosby WL, Fowke LC. ICK1, a cyclin-dependent protein kinase
inhibitor from Arabidopsis thaliana interacts with both Cdc2a and CycD3, and its expression is
induced by abscisic acid. Plant J. 1998 8; 15(4):501–10. doi: 10.1046/j.1365-313X.1998.00231.x
PMID: 9753775
102.
Fulop K, Tarayre S, Kelemen Z, Horvath G, Kevei Z, Nikovics K, et al. Arabidopsis anaphase-promot-
ing complexes: multiple activators and wide range of substrates might keep APC perpetually busy.
Cell Cycle. 2005; 4(8):1084–92. doi: 10.4161/cc.4.8.1856 PMID: 15970679
103.
Imai KK, Ohashi Y, Tsuge T, Yoshizumi T, Matsui M, Oka A, et al. The A-type cyclin CYCA2;3 is a key
regulator of ploidy levels in Arabidopsis endoreduplication. Plant Cell. 2006 2; 18(2):382–96. doi: 10.
1105/tpc.105.037309 PMID: 16415207
104.
Müssel C, Hopfensitz M, Kestler HA. BoolNet–an R package for generation, reconstruction and analy-
sis of Boolean networks. Bioinformatics. 2010 5; 26(10):1378–80. doi: 10.1093/bioinformatics/btq124
PMID: 20378558
105.
Team RC; R Foundation for Statistical Computing. R: A Language and Environment for Statistical
Computing. 2012;.
106.
Sánchez-Corrales YE, Alvarez-Buylla ER, Mendoza L. The Arabidopsis thaliana flower organ specifi-
cation gene regulatory network determines a robust differentiation process. J Theor Biol. 2010 6; 264
(3):971–83. doi: 10.1016/j.jtbi.2010.03.006 PMID: 20303988
107.
Di Cara A, Garg A, De Micheli G, Xenarios I, Mendoza L. Dynamic simulation of regulatory networks
using SQUAD. BMC Bioinformatics. 2007; 8:462. doi: 10.1186/1471-2105-8-462 PMID: 18039375
108.
Dewitte W, Riou-Khamlichi C, Scofield S, Healy JMS, Jacqmard A, Kilby NJ, et al. Altered cell cycle
distribution, hyperplasia, and inhibited differentiation in Arabidopsis caused by the D-type cyclin
CYCD3. Plant Cell. 2003 1; 15(1):79–92. doi: 10.1105/tpc.004838 PMID: 12509523
109.
Vanstraelen M, Baloban M, Da Ines O, Cultrone A, Lammens T, Boudolf V, et al. APC/C-CCS52A
complexes control meristem maintenance in the Arabidopsis root. Proc Natl Acad Sci U S A. 2009;
106(28):11806–11. doi: 10.1073/pnas.0901193106 PMID: 19553203
110.
Heyman J, De Veylder L. The anaphase-promoting complex/cyclosome in control of plant develop-
ment. Mol Plant. 2012 11; 5(6):1182–94. doi: 10.1093/mp/sss094 PMID: 23034505
111.
De Veylder L, Beeckman T, Beemster GT, de Almeida Engler J, Ormenese S, Maes S, et al. Control
of proliferation, endoreduplication and differentiation by the Arabidopsis E2Fa-DPa transcription fac-
tor. EMBO J. 2002 3; 21(6):1360–8. doi: 10.1093/emboj/21.6.1360 PMID: 11889041
112.
Poznic M. Retinoblastoma protein: a central processing unit. J Biosci. 2009 6; 34(2):305–12. doi: 10.
1007/s12038-009-0034-2 PMID: 19550046
113.
Rossignol P, Stevens R, Perennes C, Jasinski S, Cella R, Tremousaygue D, et al. AtE2F-a and AtDP-
a, members of the E2F family of transcription factors, induce Arabidopsis leaf cells to re-enter S
phase. Mol Genet Genomics. 2002 2; 266(6):995–1003. doi: 10.1007/s00438-001-0624-7 PMID:
11862494
114.
Cheng Y, Cao L, Wang S, Li Y, Shi X, Liu H, et al. Downregulation of multiple CDK inhibitor ICK/KRP
genes upregulates the E2F pathway and increases cell proliferation, and organ and seed sizes in Ara-
bidopsis. Plant J. 2013 8; 75(4):642–55. doi: 10.1111/tpj.12228 PMID: 23647236
115.
Cruz-Ramírez A, Díaz-Triviño S, Blilou I, Grieneisen VA, Sozzani R, Zamioudis C, et al. A Bistable Cir-
cuit Involving SCARECROW-RETINOBLASTOMA Integrates Cues to Inform Asymmetric Stem Cell
Division. Cell. 2012 8;.
116.
Espinosa-Soto C, Padilla-Longoria P, Alvarez-Buylla ER. A gene regulatory network model for cell-
fate determination during Arabidopsis thaliana flower development that is robust and recovers experi-
mental gene expression profiles. Plant Cell. 2004; 16(11):2923–39. doi: 10.1105/tpc.104.021725
PMID: 15486106
117.
Benítez M, Espinosa-Soto C, Padilla-Longoria P, Alvarez-Buylla ER. Interlinked nonlinear subnet-
works underlie the formation of robust cellular patterns in Arabidopsis epidermis: a dynamic spatial
model. BMC Syst Biol. 2008; 2:98. doi: 10.1186/1752-0509-2-98 PMID: 19014692
118.
Wittmann DM, Krumsiek J, Saez-Rodriguez J, Lauffenburger DA, Klamt S, Theis FJ. Transforming
Boolean models to continuous models: methodology and application to T-cell receptor signaling.
BMC Syst Biol. 2009; 3:98. doi: 10.1186/1752-0509-3-98 PMID: 19785753
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
26 / 28
119.
Krumsiek J, Pölsterl S, Wittmann DM, Theis FJ. Odefy–from discrete to continuous models. BMC Bio-
informatics. 2010; 11:233. doi: 10.1186/1471-2105-11-233 PMID: 20459647
120.
López-Avilés S, Kapuy O, Novák B, Uhlmann F. Irreversibility of mitotic exit is the consequence of sys-
tems-level feedback. Nature. 2009 5; 459(7246):592–5. doi: 10.1038/nature07984 PMID: 19387440
121.
Menges M, Samland AK, Planchais S, Murray JA. The D-type cyclin CYCD3;1 is limiting for the G1-to-
S-phase transition in Arabidopsis. Plant Cell. 2006; 18(4):893–906. doi: 10.1105/tpc.105.039636
PMID: 16517759
122.
Jurado S, Diaz-Trivino S, Abraham Z, Manzano C, Gutierrez C, del Pozo C. SKP2A, an F-box protein
that regulates cell division, is degraded via the ubiquitin pathway. Plant J. 2008; 53(5):828–41. doi:
10.1111/j.1365-313X.2007.03378.x PMID: 18036202
123.
Jurado S, Trivino SD, Abraham Z, Manzano C, Gutierrez C, Del Pozo C. SKP2A protein, an F-
box that regulates cell division, is degraded via the ubiquitin pathway. Plant Signal Behav. 2008; 3
(10):810–2. doi: 10.4161/psb.3.10.5888 PMID: 19704565
124.
Doerner P, Jorgensen JE, You R, Steppuhn J, Lamb C. Control of root growth and development by
cyclin expression. Nature. 1996; 380(6574):520–3. doi: 10.1038/380520a0 PMID: 8606769
125.
Boudolf V, Barrôco R, de Almeida Engler J, Verkest A, Beeckman T, Naudts M, et al. B1-type cyclin-
dependent kinases are essential for the formation of stomatal complexes in Arabidopsis thaliana.
Plant Cell. 2004 4; 16(4):945–55. doi: 10.1105/tpc.021774 PMID: 15031414
126.
Roeder AHK, Cunha A, Ohno CK, Meyerowitz EM. Cell cycle regulates cell type in the Arabidopsis
sepal. Development. 2012 10;.
127.
Borghi L, Gutzat R, Fütterer J, Laizet Y, Hennig L, Gruissem W. Arabidopsis RETINOBLASTOMA-
RELATED is required for stem cell maintenance, cell differentiation, and lateral organ production.
Plant Cell. 2010 6; 22(6):1792–811. doi: 10.1105/tpc.110.074591 PMID: 20525851
128.
Ferreira PC, Hemerly AS, Engler JD, van Montagu M, Engler G, Inzé D. Developmental expression of
the arabidopsis cyclin gene cyc1At. Plant Cell. 1994 12; 6(12):1763–74. doi: 10.1105/tpc.6.12.1763
PMID: 7866022
129.
Boudolf V, Vlieghe K, Beemster GT, Magyar Z, Torres Acosta JA, Maes S, et al. The plant-specific
cyclin-dependent kinase CDKB1;1 and transcription factor E2Fa-DPa control the balance of mitoti-
cally dividing and endoreduplicating cells in Arabidopsis. Plant Cell. 2004; 16(10):2683–92. doi: 10.
1105/tpc.104.024398 PMID: 15377755
130.
Li Z, Larson-Rabin Z, Masson PH, Day CD. FZR2/CCS52A1 mediated endoreduplication in Arabidop-
sis development. Plant Signal Behav. 2009 5; 4(5):451–3. doi: 10.4161/psb.4.5.8480 PMID:
19816101
131.
Takahashi N, Kajihara T, Okamura C, Kim Y, Katagiri Y, Okushima Y, et al. Cytokinins Control Endo-
cycle Onset by Promoting the Expression of an APC/C Activator in Arabidopsis Roots. Curr Biol. 2013
September; 23(18):1812–1817. doi: 10.1016/j.cub.2013.07.051 PMID: 24035544
132.
Menges M, Murray JAH. Synchronous Arabidopsis suspension cultures for analysis of cell-cycle gene
activity. Plant J. 2002 4; 30(2):203–12. doi: 10.1046/j.1365-313X.2002.01274.x PMID: 12000456
133.
Azpeitia E, Benítez M, Vega I, Villarreal C, Alvarez-Buylla ER. Single-cell and coupled GRN models of
cell patterning in the Arabidopsis thaliana root stem cell niche. BMC Syst Biol. 2010; 4:134. doi: 10.
1186/1752-0509-4-134 PMID: 20920363
134.
Barrio RA, Romero-Arias JR, Noguez MA, Azpeitia E, Ortiz-Gutiérrez E, Hernández-Hernández V,
et al. Cell patterns emerge from coupled chemical and physical fields with cell proliferation dynamics:
the Arabidopsis thaliana root as a study system. PLoS Comput Biol. 2013 5; 9(5):e1003026. doi: 10.
1371/journal.pcbi.1003026 PMID: 23658505
135.
Skirycz A, Claeys H, De Bodt S, Oikawa A, Shinoda S, Andriankaja M, et al. Pause-and-stop: the
effects of osmotic stress on cell proliferation during early leaf development in Arabidopsis and a role
for ethylene signaling in cell cycle arrest. Plant Cell. 2011; 23(5):1876–88. doi: 10.1105/tpc.111.
084160 PMID: 21558544
136.
Reyt G, Boudouf S, Boucherez J, Gaymard F, Briat JF. Iron- and Ferritin-Dependent Reactive Oxygen
Species Distribution: Impact on Arabidopsis Root System Architecture. Mol Plant. 2015; 8(3):439–
453. doi: 10.1016/j.molp.2014.11.014 PMID: 25624148
137.
Sevim V, Gong X, Socolar JES. Reliability of transcriptional cycles and the yeast cell-cycle oscillator.
PLoS Comput Biol. 2010; 6(7):e1000842. doi: 10.1371/journal.pcbi.1000842 PMID: 20628620
138.
Mu RL, Cao YR, Liu YF, Lei G, Zou HF, Liao Y, et al. An R2R3-type transcription factor gene
AtMYB59 regulates root growth and cell cycle progression in Arabidopsis. Cell Res. 2009 11; 19
(11):1291–304. doi: 10.1038/cr.2009.83 PMID: 19581938
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
27 / 28
139.
Vandepoele K, Raes J, De Veylder L, Rouze P, Rombauts S, Inze D. Genome-wide analysis of core
cell cycle genes in Arabidopsis. Plant Cell. 2002; 14(4):903–16. doi: 10.1105/tpc.010445 PMID:
11971144
140.
Gutierrez C. The Arabidopsis cell division cycle. Arabidopsis Book. 2009; 7:e0120. doi: 10.1199/tab.
0120 PMID: 22303246
141.
Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer.
2009; 9(3):153–66. doi: 10.1038/nrc2602 PMID: 19238148
142.
Wittenberg C, Reed SI. Cell cycle-dependent transcription in yeast: promoters, transcription factors,
and transcriptomes. Oncogene. 2005 4; 24(17):2746–55. doi: 10.1038/sj.onc.1208606 PMID:
15838511
143.
Bertoli C, Skotheim JM, de Bruin RAM. Control of cell cycle transcription during G1 and S phases. Nat
Rev Mol Cell Biol. 2013 8; 14(8):518–28. doi: 10.1038/nrm3629 PMID: 23877564
144.
Breeden LL. Periodic transcription: a cycle within a cycle. Curr Biol. 2003 1; 13(1):R31–8. doi: 10.
1016/S0960-9822(02)01386-6 PMID: 12526763
145.
Cokus S, Rose S, Haynor D, Grønbech-Jensen N, Pellegrini M. Modelling the network of cell cycle
transcription factors in the yeast Saccharomyces cerevisiae. BMC Bioinformatics. 2006; 7:381. doi:
10.1186/1471-2105-7-381 PMID: 16914048
146.
Francis D. A commentary on the G2/M transition of the plant cell cycle. Ann Bot. 2011 5; 107(7):1065–
70. doi: 10.1093/aob/mcr055 PMID: 21558458
147.
Shimotohno A, Ohno R, Bisova K, Sakaguchi N, Huang J, Koncz C, et al. Diverse phosphoregulatory
mechanisms controlling cyclin-dependent kinase-activating kinases in Arabidopsis. Plant J. 2006; 47
(5):701–10. doi: 10.1111/j.1365-313X.2006.02820.x PMID: 16856985
148.
Cook GS, Grønlund AL, Siciliano I, Spadafora N, Amini M, Herbert RJ, et al. Plant WEE1 kinase is cell
cycle regulated and removed at mitosis via the 26S proteasome machinery. J Exp Bot. 2013; 64
(7):2093–2106. doi: 10.1093/jxb/ert066 PMID: 23536609
149.
Dissmeyer N, Weimer AK, Pusch S, De Schutter K, Alvim Kamei CL, Nowack MK, et al. Control of cell
proliferation, organ growth, and DNA damage response operate independently of dephosphorylation
of the Arabidopsis Cdk1 homolog CDKA;1. Plant Cell. 2009; 21(11):3641–54. doi: 10.1105/tpc.109.
070417 PMID: 19948791
150.
Dissmeyer N, Weimer AK, De Veylder L, Novak B, Schnittger A. The regulatory network of cell-cycle
progression is fundamentally different in plants versus yeast or metazoans. Plant Signal Behav. 2010;
5(12):1613–8. doi: 10.4161/psb.5.12.13969 PMID: 21139435
Dynamic Gene Regulatory Network Model of A. thaliana Cell Cycle
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004486
September 4, 2015
28 / 28
|
26340681
|
MYB77 = ( E2Fb AND ( ( ( NOT RBR ) ) OR ( ( CYCD3;1 ) AND ( ( ( NOT KRP1 ) ) ) ) ) )
E2Fe = ( ( CYCD3;1 AND ( ( ( KRP1 ) AND ( ( ( NOT E2Fc AND NOT RBR AND NOT MYB77 AND NOT E2Fb ) ) ) ) ) ) OR ( RBR AND ( ( ( KRP1 ) AND ( ( ( NOT E2Fc AND NOT MYB77 AND NOT CYCD3;1 AND NOT E2Fb ) ) ) ) ) ) OR ( E2Fb AND ( ( ( RBR AND KRP1 ) AND ( ( ( NOT E2Fc AND NOT MYB77 AND NOT CYCD3;1 ) ) ) ) ) ) OR ( KRP1 AND ( ( ( NOT E2Fc AND NOT RBR AND NOT MYB77 AND NOT CYCD3;1 AND NOT E2Fb ) ) ) ) OR ( MYB77 ) ) OR NOT ( E2Fc OR RBR OR MYB77 OR CYCD3;1 OR KRP1 OR E2Fb )
KRP1 = ( ( MYB3R1/4 ) AND NOT ( CDKB1;1 AND ( ( ( SCF AND CYCA2;3 ) ) ) ) ) OR ( ( MYB77 ) AND NOT ( CDKB1;1 AND ( ( ( SCF AND CYCA2;3 ) ) ) ) )
E2Fc = ( ( MYB3R1/4 ) AND NOT ( SCF AND ( ( ( CYCD3;1 ) AND ( ( ( NOT KRP1 ) ) ) ) ) ) ) OR ( ( ( E2Fa ) AND NOT ( SCF AND ( ( ( CYCD3;1 ) AND ( ( ( NOT KRP1 ) ) ) ) ) ) ) AND NOT ( RBR ) )
MYB3R1/4 = ( ( MYB3R1/4 AND ( ( ( CYCB1;1 ) ) ) ) AND NOT ( KRP1 ) ) OR ( MYB77 )
E2Fb = ( ( E2Fa ) AND NOT ( RBR ) )
SCF = ( ( MYB3R1/4 ) AND NOT ( APC/C ) ) OR ( ( E2Fb AND ( ( ( CYCD3;1 ) AND ( ( ( NOT KRP1 ) ) ) ) OR ( ( NOT RBR ) ) ) ) AND NOT ( APC/C ) )
CDKB1;1 = ( MYB3R1/4 ) OR ( MYB77 ) OR ( ( E2Fb AND ( ( ( CYCD3;1 ) AND ( ( ( NOT KRP1 ) ) ) ) OR ( ( NOT RBR ) ) ) ) AND NOT ( E2Fc ) )
RBR = ( ( E2Fa AND ( ( ( NOT CYCD3;1 ) ) OR ( ( KRP1 ) ) ) ) AND NOT ( RBR AND ( ( ( KRP1 ) ) OR ( ( NOT CYCD3;1 ) ) ) ) ) OR ( MYB3R1/4 AND ( ( ( NOT CYCD3;1 ) ) OR ( ( KRP1 ) ) ) )
CYCD3;1 = NOT ( ( SCF ) )
CYCB1;1 = ( ( MYB77 ) AND NOT ( APC/C ) ) OR ( ( MYB3R1/4 ) AND NOT ( APC/C ) ) OR ( ( ( E2Fb AND ( ( ( CYCD3;1 ) AND ( ( ( NOT KRP1 ) ) ) ) OR ( ( NOT RBR ) ) ) ) AND NOT ( E2Fc ) ) AND NOT ( APC/C ) )
E2Fa = ( ( ( E2Fa ) AND NOT ( CDKB1;1 AND ( ( ( CYCA2;3 ) ) ) ) ) OR ( CDKB1;1 AND ( ( ( NOT E2Fc AND NOT E2Fa AND NOT CYCA2;3 ) ) ) ) OR ( CYCA2;3 AND ( ( ( NOT CDKB1;1 AND NOT E2Fc AND NOT E2Fa ) ) ) ) ) OR NOT ( CDKB1;1 OR E2Fc OR E2Fa OR CYCA2;3 )
CYCA2;3 = ( ( MYB3R1/4 ) AND NOT ( APC/C ) ) OR ( ( MYB77 ) AND NOT ( APC/C ) )
APC/C = ( ( MYB3R1/4 ) AND NOT ( E2Fe ) ) OR ( ( ( E2Fa ) AND NOT ( RBR ) ) AND NOT ( E2Fe ) ) OR ( ( MYB77 ) AND NOT ( E2Fe ) )
|
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
DOI 10.1186/s12976-015-0011-4
RESEARCH ARTICLE
Open Access
Fanconi anemia cells with unrepaired DNA
damage activate components of the
checkpoint recovery process
Alfredo Rodríguez1,2, Leda Torres1, Ulises Juárez1, David Sosa1, Eugenio Azpeitia3,4,5,
Benilde García-de Teresa1, Edith Cortés6, Rocío Ortíz6, Ana M. Salazar7, Patricia Ostrosky-Wegman7,
Luis Mendoza4,7
† and Sara Frías1,7*†
*Correspondence:
sarafrias@biomedicas.unam.mx
†Equal contributors
1Laboratorio de Citogenética,
Departamento de Investigación en
Genética Humana, Instituto
Nacional de Pediatría, D.F., México
7Instituto de Investigaciones
Biomédicas, Universidad Nacional
Autónoma de México, D.F., México
Full list of author information is
available at the end of the article
Abstract
Background: The FA/BRCA pathway repairs DNA interstrand crosslinks. Mutations in
this pathway cause Fanconi anemia (FA), a chromosome instability syndrome with bone
marrow failure and cancer predisposition. Upon DNA damage, normal and FA cells
inhibit the cell cycle progression, until the G2/M checkpoint is turned off by the checkpoint
recovery, which becomes activated when the DNA damage has been repaired.
Interestingly, highly damaged FA cells seem to override the G2/M checkpoint. In this
study we explored with a Boolean network model and key experiments whether
checkpoint recovery activation occurs in FA cells with extensive unrepaired DNA damage.
Methods: We performed synchronous/asynchronous simulations of the FA/BRCA
pathway Boolean network model. FA-A and normal lymphoblastoid cell lines were used
to study checkpoint and checkpoint recovery activation after DNA damage induction.
The experimental approach included flow cytometry cell cycle analysis, cell division
tracking, chromosome aberration analysis and gene expression analysis through qRT-PCR
and western blot.
Results: Computational simulations suggested that in FA mutants checkpoint recovery
activity inhibits the checkpoint components despite unrepaired DNA damage, a
behavior that we did not observed in wild-type simulations. This result implies that FA
cells would eventually reenter the cell cycle after a DNA damage induced G2/M
checkpoint arrest, but before the damage has been fixed. We observed that FA-A cells
activate the G2/M checkpoint and arrest in G2 phase, but eventually reach mitosis and
divide with unrepaired DNA damage, thus resolving the initial checkpoint arrest. Based
on our model result we look for ectopic activity of checkpoint recovery components.
We found that checkpoint recovery components, such as PLK1, are expressed to a
similar extent as normal undamaged cells do, even though FA-A cells harbor highly
damaged DNA.
Conclusions: Our results show that FA cells, despite extensive DNA damage, do not
loss the capacity to express the transcriptional and protein components of checkpoint
recovery that might eventually allow their division with unrepaired DNA damage. This
might allow cell survival but increases the genomic instability inherent to FA
individuals and promotes cancer.
Keywords: DNA damage, Checkpoint recovery, Boolean network model
© 2015 Rodríguez et al. Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://
creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 2 of 22
Introduction
The molecular basis of the DNA damage response (DDR) has been largely elucidated
through the study of the rare chromosome instability syndromes (CIS) [1] which are
cytogenetically characterized by the spontaneous appearance of chromosome aberrations
(CA) as well as hypersensitivity to specific DNA damaging agents [2–4]. The best-known
CIS include Bloom syndrome (BS) which appears due to mutations in BLM helicase [5, 6]
and results in increased sister chromatid exchanges [7], Ataxia Telangiectasia (AT) that
shows particular clonal chromosome rearrangements as a consequence of mutations in
the checkpoint kinase ATM gene[8–11], and Fanconi anemia (FA) [12] whose phenotype
results from mutations in any of the genes that conform the FA/BRCA pathway [13–19]
and consists of chromatidic breaks, iso-chromatidic breaks and radial exchange figures
among chromosomes. Even if these breaks and radials are predominantly seen in FA, they
can also be observed in BS and AT [4, 20]. Although patients affected by CIS display phe-
notypic similarities, such as growth defects, compromised immunological system and an
increased risk to develop cancer [1, 20], each syndrome presents particular phenotypes
and pivotal data. Namely, BS shows sun sensitivity [5], AT presents progressive cerebel-
lar ataxia and oculo-cutaneous telangiectases [8], while FA is characterized by congenital
malformations and progressive bone marrow failure [21]. The products of these genes
interact in the cell’s DNA damage response [1], and thus the deficiency of any of these
proteins diminishes the efficiency of a cell to cope with DNA damage, leading to their
accumulation.
Given the critical role that these proteins have in the protection of the human genome,
certain authors have speculated that survival of CIS patients is an oddity and that
cells escaping apoptotic death do so by constitutively inducing alternative replication or
DNA damage tolerance pathways, which might contribute to the characteristic mutator
phenotypes observed in the CIS [22].
In the particular case of FA, cells are hypersensitive to agents that create DNA inter-
strand crosslinks (ICL), such as mitomycin C (MMC) or diepoxybutane (DEB) [21]. The
treatment of FA cells with MMC or DEB induces a blockage during the G2 phase of
the cell cycle and exacerbates the frequency of CAs, including double strand breaks
(DSBs) and radial exchange figures [23]. Biallelic mutations in at least one of 18 distinct
FANC genes can generate FA. The products of these genes interact in the so-called Fan-
coni Anemia/Breast Cancer (FA/BRCA) pathway [13–18], involved in the repair of the
DNA damage generated by intrinsic acetaldehydes and extrinsic ICL inducing agents.
Therefore, a deficiency in this pathway results in DNA damage accumulation that might
originate congenital malformations, uncontrolled hematopoietic cell death and cancer in
FA patients [24–27].
Over the years, the FA diagnosis assays and experimental approaches have shown that
a great proportion of FA cells succumb to DNA damage due to their inherent repair defi-
ciencies. However, some cells are able to tolerate high levels of DNA damage and progress
into mitosis despite a great amount of CAs. The mechanisms that allow the cells with
CAs to omit the DNA damage integrity checkpoints remain uncertain because the more
obvious candidate, the G2/M checkpoint, is considered to be properly activated in FA
cells [28–30]. Thus, the idea of a malfunctioning checkpoint in FA cells has been ruled
out and it is presumed that some other mechanisms are responsible for the checkpoint
override in FA cells with unrepaired DSBs.
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 3 of 22
In recent times, an attenuated G2 checkpoint phenotype, characterized by low levels
of CHK1 (NP_001107594.1) and p53 (NP_000537.3), absence of the G2 phase arrest, and
arrival to metaphase with a large number of MMC-induced CAs has been described
in cells from adult FA individuals [31]. It has been suggested that the G2 checkpoint
attenuation could be an important contributor for the increased life expectancy of these
FA patients, and that the release of cells with unrepaired DSBs could promote neoplas-
tic transformation [31]. Nevertheless, since non-attenuated FA cells carrying unrepaired
DNA damage achieve a correct G2/M checkpoint activation [28–30], the aforementioned
mechanism seems to be a particular scenario rather than a general mechanism to enable
the resolution of the G2 checkpoint blockage.
Network modeling has been previously used with success to study the dynamics of
biological systems [32–37]. Particularly, we developed a Boolean network model (BNM)
for the FA/BRCA pathway [38], in which we observed that the inclusion of the check-
point recovery (CHKREC) node is crucial for the network correct function. In our model,
the CHKREC node represents the process that relieves the inhibition of the checkpoint
machinery over the mitosis-promoting factor (Cyclin B/CDK1) after a complete DNA
damage repair to allow further cell division [39–42]. This node comprises the G2 tran-
scriptional program that activates the expression of genes driving the G2/M transition
and the protein program that inactivates the γ H2AX histone (NP_002096.1) and check-
point kinases [43]. We presumed that CHKREC activation might be releasing cells with
unrepaired DNA damage in FA mutants. To test this possibility, as well as to validate
the inclusion of the node itself in the FA/BRCA network, we used a simplified version
of our previously published FA/BRCA pathway BNM and experimentally determined if
CHKREC components become activated in FA cells during G2/M during MMC-induced
arrest.
Materials and methods
Model and simulations
The simplification of the FA/BRCA network was done by reorganizing the existing 28
nodes and 122 interactions [38], resulting in a deterministic BNM with 15 nodes and 66
interactions (Fig.1 and Table 1) that vastly simplifies the computational analysis while
maintaining the qualitative dynamical behavior of the original FA/BRCA network. The
simplification was made by collapsing the network components that share functions or
belong to a single pathway into one single node. We were careful to preserve all the impor-
tant functional categories of the network and made sure to recover the behavior of the
wild type and mutant networks. The modifications and simplification criteria are listed in
Table 2.
Simulations were performed for the wild type and all possible gain of function or null
mutants of the model with synchronous and asynchronous update regimes. Here we
report the simulations exploring checkpoint and CHKREC function in wild type and FA
core mutants. These null mutants were simulated fixing to zero the node of interest.
In these mutants we simulated the response to ICLs, whose presence is dependent on
the time that the system requires to turn it off. With our model we simulated two biolog-
ically relevant conditions, a short exposure to ICLs, which is supposed to be repaired fast
and efficiently by the FA/BRCA pathway (Fig. 2b) and a persistent exposure to damaging
agents, which is more difficult to face given the accumulation of damage and saturation
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 4 of 22
Fig. 1 (See legend on next page)
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 5 of 22
(See figure on previous page)
The latest FA/BRCA network. In response to an ICL, the FA/BRCA network responds by blocking the cell cycle
through the ATR and ATM checkpoint kinases and their downstream target p53. Similarly, the FA core
complex (FAcore) becomes activated and ubiquitinates FANCD2I complex, which in turn recruits DNA
endonucleases (NUC1 and NUC2). These endonucleases unhook the ICL generating a DNA adduct (ADD) and
a double strand break (DSB). Translesion synythesis (TLS) takes over the ADD while the DSB can be rejoined
either by FA/BRCA-dependent Homologous Recombination (FAHRR), FA/BRCA-independent Homologous
Recombination (HRR2), or by the error prone Non-Homologous End-Joining (NHEJ) pathways. Finally, we
predict that the CHKREC node, composed by the G2/M transcriptional program and checkpoint recovery
proteins, turns off the checkpoint and DNA repair proteins. Rectangles represent proteins or protein
complexes, pointed arrows are positive regulatory interactions, and dashed lines with blunt arrows are
negative regulatory interactions. Readers may refer to [38] for a more detailed description of the FA/BRCA
pathway
of the DNA repair pathway (Fig. 2c). The response to short ICL exposure was simulated
in both the wild type (Fig. 2b) and FAcore mutant (Fig. 2d) with the ICL value ON only
at the starting time step; whereas a continuous exposure to DNA damage was simulated
fixing the ICL value to 1 during the entire simulation. We performed exhaustive searches
of all possible trajectories and attractors in the system.
Implementation
The current FA/BRCA network is available through the supplementary file FAnetwork.r
this file has been tested using R (v3.1.1) package BoolNet (v1.63) [66]. Additionally, the
SBML-qual implementation of the model obtained by using the toSBML() function of
BoolNet is provided as the supplementary file FAnetwork.sbml. The generated file was
validated using the online service at http://sbml.org/Facilities/Validator/.
Table 1 Boolean functions for the nodes in the FA/BRCA network
RULES
REFERENCES
ICL ←ICL ∧¬ DSB
[38]
FAcore ←ICL ∧(ATR ∨ATM) ∧¬ CHKREC
[14, 16, 44–46]
FANCD2I ←FAcore ∧((ATR ∨ATM) ∨((ATR ∨ATM) ∧DSB)) ∧
[47–49]
¬ (CHKREC)
NUC1 ←ICL ∧FANCD2I
[50]; [38]
NUC2 ←(ICL ∧(ATR ∨ATM) ∧¬ (FAcore ∧FANCD2I)) ∨
[51]; [38]
(ICL ∧NUC1 ∧p53 ∧¬(FAcore ∧FANCD2I))
ADD ←(NUC1 ∨NUC2 ∨(NUC1 ∧NUC2)) ∧¬ (TLS)
[47, 50, 51]
DSB ←(NUC1 ∨NUC2) ∧¬ (NHEJ ∨FAHRR ∨HRR2)
[50, 52]
TLS ←(ADD ∨(ADD ∧FAcore)) ∧¬ (CHKREC)
[53, 54]
FAHRR ←DSB ∧FANCD2I ∧¬ (NHEJ ∧CHKREC)
[53, 54]
HRR2 ←(DSB ∧NUC2 ∧NHEJ ∧ICL ∧¬ (FAHRR ∨CHKREC)) ∨
[38]
(DSB ∧NUC2 ∧TLS ∧¬ (NHEJ ∨FAHRR ∨CHKREC))
NHEJ ←(DSB ∧NUC2 ∧¬ (FAHRR ∨HRR2 ∨CHKREC))
[49, 52, 55–57]
ATR ←(ICL ∨ATM) ∧¬ CHKREC
[58–60]
ATM ←(ATR ∨DSB) ∧¬ CHKREC ∨FAcore
[61, 62]
P53 ←((ATR ∨ATM) ∨NHEJ) ∧¬ CHKREC
[58, 63, 64]
CHKREC ←((TLS ∨NHEJ ∨FAHRR ∨HRR2) ∧¬ DSB ) ∨
[52, 53];
((¬ ADD) ∧(¬ ICL) ∧(¬ DSB) ∧¬ (CHKREC))
[38, 65]
Key references are included. Full discussion of interactions can be found in [38]
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 6 of 22
Table 2 Network model simplification criteria
Node in the
Nodes in the
Simplification criteria
original BNM
simplified BNM
ICL
ICL
Unchanged node
FANCM, FAcore
FAcore
ICL recognition proteins working
together in the upstream FA/BRCA
pathway
FANCD2I
FANCD2I
Unchanged node
MUS81
NUC1
Nuclease mediated
ICL incision
XPF, FAN1
NUC2
Nuclease mediated
ICL incision
ADD
ADD
Unchanged node
DSB
DSB
Unchanged node
ATR, CHK1, H2AX
ATR
These proteins act in the
Checkpoint pathway
ATM, CHK2, H2AX
ATM
These proteins act in the
Checkpoint pathway
p53
p53
Unchanged node
PCNATLS
TLS
This is only a change in name
FANCJMLH1, MRN, BRCA1,
FAHRR
These proteins act in the
FANCD1N, RAD51, HRR,
Homologous Recombination
ssDNARPA
Repair pathway
——
HRR2
New node representing the
alternative Homologous
Recombination Repair Pathway
KU, DNAPK, NHEJ
NHEJ
These proteins act in the
Non-Homologous End-Joining
DNA repair pathway
USP1, CHKREC
CHKREC
Global negative regulators
of the FA/BRCA pathway
Cell culture and treatments
Lymphoblastoid cell lines from FA-A VU817 (kindly donated by Dr. Hans Joenje, VU
University Medical Center) and normal NL-49 (generated in our institution under writ-
ten informed consent) were maintained in RPMI 1640 medium supplemented with 10 %
fetal calf serum, 1 % non-essential aminoacids and 1 % sodium pyruvate (all from GIBCO,
Waltham, Massachusetts, USA). During experiments 300,000 cell/ml were exposed to 10
ng/ml of MMC (Sigma-Aldrich Co, St. Louis MO, USA) for 24 h and harvested to evaluate
different markers. All the experiments were run by triplicate.
Chromosome aberration and nuclear division index analysis
For chromosome aberration analysis, colchicine (Sigma-Aldrich Co, St. Louis MO, USA)
(final concentration of 0.1 μg/ml) was added to cell cultures one hour before harvesting
with the conventional method. Twenty five metaphases per experimental condition were
scored by recording the number of chromatid breaks, chromosome breaks and radial
figures. A cytokinesis block assay, using 3 μg/ml of cytochalasin B (Sigma-Aldrich Co,
St. Louis MO, USA), was implemented to obtain binucleated and tetranucleated cells:
after exposing the cells to MMC for 24 h, they were washed, reincubated with fresh
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 7 of 22
A
B
C
D
E
Fig. 2 FA network simulations. a The current information regarding the FA/BRCA pathway have not
uncovered the mechanism that allows the resolution of the G2/M checkpoint after DNA damage and further
cell division. b Trajectories and attractor of the wild type FA/BRCA network under an ICL pulse. In this
simulation wild type cells repair DNA damage through the FA/BRCA pathway and arrive to CCP attractor after
activating the CHKREC node once the damage has been fixed. The inclusion of the CHKREC node, as a
checkpoint negative regulator, allows to explore the mechanisms behind cell division after checkpoint
resolution. c In response to a continuous ICL DNA damage, wild type cells arrive to a CCA attractor with
activation of the checkpoint and DNA damage repair nodes,the CHKREC node becomes eventually activated
in this attractor. d Under and ICL pulse FAcore mutant cells activate the NHEJ pathway to repair DNA damage
and arrive to a CCP attractor. e In response to a continuous ICL DNA damage, FAcore mutant cells
concomitantly activate the checkpoint and the CHKREC nodes. Node names are indicated at the topmost
row. The leftmost column indicates simulation time steps in arbitrary units. Time steps corresponding to
trajectories are indicated and time steps corresponding to attractors are indicated by shaded gray and “ATT”.
For illustrative purpose cyclic attractors are represented twice
cytochalasin B for another 24 h and harvested using a 7:1 methanol:acetic acid fixative.
Five hundred cells were scored to quantify the number of micronuclei, mononucleated,
binucleated and tetranucleated cells in every experimental condition [67].
Flow cytometry analysis
To determine cell cycle distribution and mitotic index the cells were fixed with 70 %
ice-cold ethanol, washed twice with PBS (GIBCO, Waltham, Massachusetts, USA) and
permeabilized with 0.1 % PBS 1X + Triton X100. The MPM2 antibody (CellSignaling,
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 8 of 22
Boston MA, USA) was used to determine the number of cells in M phase. The antibody
was marked with the labeling anti-mouse Alexa-Fluor 488 fluorophore from the Zenon
Tricolor Mouse IgG Labeling Kit # 1 (Invitrogen, Carlsbad, CA, USA) according to man-
ufacturer instructions. The cells were incubated during 1 h with the antibody, washed
with PBS/NGS 10 % and counterstained with propidium iodide (Sigma-Aldrich Co, St.
Louis MO, USA). A total of 20,000 events were scored in a FACSCan (Beckton Dickinson,
Ontario, CA) cytometer and the analysis was performed using the CellQuest program
version 3.2.1.
RNA extraction and quantitative real-time PCR (qRT-PCR)
Total RNA was obtained employing the combined method of TRIzol (Invitrogen,
Carlsbad, CA, USA) followed by RNeasy mini procedure (Qiagen, Valencia, CA, USA),
according to manufacturer instructions. Before retro-transcription, 1 μg of total RNA
was treated with 0.1 U RNase-free DNase I (Invitrogen, Carlsbad, CA, USA) in 20
mM Tris-HCl, pH 8.3, 50 mM KCl, and 1 mM MgCl2 for 15 min at room tempera-
ture. The enzyme was inactivated by adding EDTA to a final concentration of 1 mM
followed by incubation at 65°C/10 min. Total RNA was retro-transcribed into cDNA
using the Transcriptor First Strand cDNA Synthesis Kit (Roche Diagnostics, GmbH,
Mannheim, Germany) using anchored-oligo (dT) 18 primer (50-pmol/μL) and Random
hexamer primer (600 pmol/μL), protector RNase Inhibitor (20 U), and Transcriptor
Reverse Transcriptase (10 U). Total RNA and cDNA were quantified using a Nanodrop
ND 1000 spectrophotometer (Nanodrop Technologies, Wilmington, DE, USA). Real-time
quantitative-PCR (qRT-PCR) was performed by duplicate for each cell line, treatment
and biological repeat using 2 μg of cDNA per reaction with the Universal Probes sys-
tem (Roche Diagnostics, GmbH, Mannheim, Germany) and the Light Cycler Taq Man
Master kit (Roche Diagnostics, GmbH, Mannheim, Germany). 7SL (NR_002715.1), β2
microglobulin (NM_004048.2) and β-actin (NM_001017992.3) gene expression were
used as reference. Primers for each gene were designed on-line with the ProbeFinder
Software (http://www.universalprobelibrary.com) and manufactured by the Sequencing
and Synthesis Unit (IBT, UNAM). The qRT -PCR was carried out in a Light Cycler 2.0
Carousel Roche equipment.
Protein extraction and immunoblot
Cells were harvested in TLB lysis buffer supplemented with the Complete C protease
and phosphatase inhibitors mix (Roche, Mannheim, Germany). Quantification was made
with Bradford ready to use reagent (Biorad, Hercules, CA). Total cell protein (10μg) was
separated by 12 % SDS- PAGE, transferred to nitrocellulose membrane (Biorad, Hercules,
CA) and incubated with primary antibodies overnight at 4°C followed by incubation with
goat-anti-mouse (Invitrogen, Carlsbad, CA, USA) or goat-anti-rabbit (Invitrogen, Carls-
bad, CA, USA) HRP tagged secondary antibodies. Bands were visualized with Lumigen
on Amersham Hyperfilm (GE Healthcare, Fairfield, CT, USA). Primary antibodies used
are listed below: anti-WEE1 (NP_001137448.1) (Abcam, Cambridge, UK), anti-WIP1
(NP_003611.1 ) (Abcam, Cambridge, UK), anti-pCHK1 Ser345 (Cell Signaling, Boston
MA, USA), anti-γ H2AX (Genetex, Irvine, CA), anti-p21 (NP_000380.1) (Genetex, Irvine,
CA), anti-MYT1 (NP_004526.1) (Genetex, Irvine, CA), anti-Aurora A (NP_003591.2)
(Abcam, Cambridge, UK), anti-CDC25B (NP_001274445.1) (Genetex, Irvine, CA) and
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 9 of 22
anti-PLK1 (NP_005021.2) (Abcam, Cambridge, UK); anti-GAPDH (NP_001243728.1)
was used as loading control (Genetex, Irvine, CA).
Statistical analysis
Experimental groups were compared using two way ANOVA, followed by Tukey’s post-
hoc test. A difference was considered significant if p < 0.05.
Results
FA/BRCA network analyses show that CHKREC promotes cell division in FA mutants with
DNA damage
Appropriate function of the FA/BRCA pathway guarantees the complete repair of ICLs
and correct checkpoint activation impedes cell division upon DNA damage detection
[68]. Therefore an accurate model of the FA/BRCA pathway should show cell division
after complete DNA damage repair in wild-type cells. In our previous work [38], we
demonstrated that the inclusion of the CHKREC node is crucial to reproduce correctly
the DNA repair behavior. Without CHKREC, as a negative regulator of the checkpoint
nodes, the network remains in a permanent arrest after DNA repair (Fig. 2a). Hence,
CHKREC provides a mechanism that allows the cell to resolve the checkpoint (Fig. 2b).
We performed synchronous and asynchronous simulations with the updated and sim-
plified version of the FA/BRCA network and observed that the simplified model is able
to reproduce all the previously reported results (Fig. 2b and data not shown). Only
synchronous simulations are shown given that asynchronous update results in complex
trajectories, while preserving the attractors of the original model [38]. Hence, we decided
to use our new version of the network model to deeply study the role of CHKREC in the
abnormal behavior of FA cells.
Ninety percent of FA patients carry mutations in one of the components of the FA
core complex, including FANCA (NP_000126.2), FANCB (NP_001018123.1), FANCC
(NP_000127.2), FANCE (NP_068741.1), FANCF (NP_073562.1), FANCG (NP_004620.1),
FANCL (NP_001108108.1) and FANCM (NP_001295063.1) [21]. Hence, to study the role
of CHKREC in FA cells, we simulated the FA core complex mutant, represented in our
model by the FA core node, and compared its dynamic behavior to a wild type network.
Our simulations recapitulate two cellular behaviors relevant to DNA damage that are
represented by two specific attractors. We denominated them as the cell cycle progression
attractor (CCP), and the cell cycle arrest attractor (CCA). The CCP attractor is charac-
terized by the CHKREC-mediated inactivation of every checkpoint node, namely ATM,
ATR and p53, followed by CHKREC oscillations. It has been experimentally proven that
CHKREC is required for the activation of the genes and proteins that release the G2/M
checkpoint to allow cell cycle progression [39, 41–43]. Hence, the cyclic behavior of the
CHKREC node in the CCP attractor represents the periodical transition into the cell
cycle, and should ideally be reached when DNA damage has been repaired. In our simu-
lations both wild type and FA core mutant reach the CCP attractor after an ICL pulse of
damage (Fig. 2b,d).
On the other hand, CCA is a cyclic attractor that represents a checkpoint mediated
cell cycle arrest that is reached when DNA damage persists and the cell is engaged in
a DNA repair process. Once the system has reached CCA there is recurrent activation
of the DNA damage repair and the checkpoint nodes, accompanied by CHKREC node
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 10 of 22
activation, thus CHKREC activation might occur during an ongoing CCA but the cell
would not divide unless the checkpoint nodes are turned off, which in turn would not
occur until the DNA damage has been completely removed. Although more than one
combination of node activation patterns can be interpreted as a CCA attractor, all such
patterns share the activation of the DNA damage and the checkpoint nodes followed by
activation of CHKREC.
In our simulations with a constant ICL damage the wild type (Fig. 2c) and FA core
mutant (Fig. 2e) networks reach a CCA attractor with checkpoint and CHKREC activa-
tion. In the wild-type simulation we observe that the checkpoint components are never
completely down-regulated in presence of DSBs or during the ICL stimulus, however the
FA core mutants have a transient state in which DSBs are activated and the checkpoint
components are inactivated, as a response to CHKREC activation in the previous step.
This result suggests that FA cells might overcome, through CHKREC activation, the cell
cycle arrest despite unrepaired DNA damage.
Our modeling approach has advanced many interesting predictions about the effect of
FA mutations during the DNA repair process. In the next section we focused on the one
that we considered more general and important. Namely, that CHKREC inhibition over
the checkpoint components might allow the division of FA cells even if DNA has not been
completely repaired. Hence, we verified if CHKREC activation might occur in FA cells
after DNA damage induction allowing their eventual cell division, even in the presence of
unrepaired DNA damage.
CHKREC components are activated in FA cells with unrepaired DNA damage
Cells should divide only after successful and thorough DNA repair [68, 69], which is
achieved through efficient DNA repair and accurate G2/M checkpoint activation. FA core
mutants are DNA repair deficient but G2/M checkpoint proficient, therefore the fact
that they are able to divide despite a strong G2/M checkpoint activation and carrying
unrepaired DNA damage is remarkable. Our BNM anticipates that turning off a DNA
damage-induced G2/M checkpoint might occur through CHKREC activation, thus allow-
ing cell division. We verified this prediction by following the transit through G2 and M
phases in the presence of DNA damage in wild type (NL49) and FA-A (VU817) cell lines
exposed to MMC.
First, we evaluated checkpoint activation using several markers. Using PI cell cycle flow
cytometry analysis we observed that treatment with MMC induces an over time increase
in the number of FA-A cells arrested in G2 when compared to normal cells (Fig. 3a, left
panel), as well as a reduction in the number of mitotic cells (MPM2+ cells in Fig. 3a, right
panel), accompanied by a lag of approximately 6–12 hours in the peak of MPM2+ cells in
both MMC-treated FA and normal cells, compared to their respective untreated controls
(see 24 and 30 hrs of MMC treatment). However normal MMC treated cells have the
highest peak as they deliver a bigger number of cells into M phase. This lag might indicate
that, while repairing the MMC-induced DNA damage, the cells postpone the resolution
of the G2 checkpoint. This arrest is shorter in normal cells given that they repair in a
more efficient way thus having a more prominent contribution to the mitotic index when
compared to FA cells. The highest percentage of MPM2+ cells of MMC treated normal
cells might indicate a sharp delivery of previously G2 arrested cells, contrary to a smooth
delivery of untreated normal cells.
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 11 of 22
A
B
C
Fig. 3 FA cells arrest at G2 phase in response to MMC. a Flow cytometry analysis showing accumulation of
FA cells in G2 in response to MMC (left panel) and diminished number of FA mitotic cells compared to
normal cells (right panel). b FA cells activate CHK1 kinase in response to MMC treatment. c FA cells increase
the expression of p21 mRNA as showed by qRT-PCR analysis (n = 3 independent experiments, p < 0.05)
In FA-A cells treated with MMC we also observed increased CHK1 phosphoryla-
tion (Fig. 3b), a classical checkpoint activation marker, along with increased p21 mRNA
expression (Fig. 3c). p21 is the main p53 target and is an important player for cell cycle
arrest. The expression of this gene shows that the cell is committed to cell cycle arrest
and its continuous expression is necessary to prevent cell division in cells that carry unre-
paired chromosomes [68, 69]. These experiments show that FA-A cells are able to activate
mechanisms that halt cell cycle progression at the G2 phase upon DNA damage induction.
We then evaluated if FA-A and normal cells were able to divide despite unrepaired DNA
damage. We quantified the cell division capacity after MMC treatment by performing a
cytokinesis block assay with cytochalasin B (CB). Meanwhile, the DNA damage was eval-
uated by recording the frequency of micronuclei in multinucleated cells and the frequency
of CAs in metaphase spreads.
CB experiments showed that treatment with MMC increased the proportion of
mononucleated cells (cells that still do not divide due to G2 halt) (Fig. 4a upper panel),
while the number of binucleated cells irrespective of the cell type (NL49 or VU817) or the
addition or not of MMC remained the same (Fig. 4a middle panel). Remarkably, MMC
treatment reduced significantly the number of tetranucleated FA cells (Fig. 4a bottom
panel). On the other hand, the analysis of metaphase spreads showed that FA-A cells
reached mitosis with a significantly higher frequency of CAs (Fig. 4b upper panel) than
normal cells, and were able to divide despite unrepaired DNA damage, i.e. micronuclei
(Fig. 4b bottom panel). These experiments show that FA-A cells first arrest in response to
DNA damage but eventually reach cell division regardless of CA.
As suggested by our model, CHKREC activation could be relieving cell cycle arrest
mediators, leading the cell to divide. To determine if the CHKREC components became
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 12 of 22
A
B
Fig. 4 FA cells divide despite MMC treatment and cell cycle arrest. a The number of mononucleated cells
(upper panel), binucleated cells (middle panel) and tetranucleated cells (bottom panel) was quantified after
exposure to MMC (24 h) and Citochalasin B (48 h). The number of basal binucleated and tetranucleated cells
was counted without Citochalasin B treatment and rested from the total number (data not shown). b Despite
G2 arrest FA cells arrive to mitosis and divide with unrepaired DNA damage as demonstrated by two
independent DNA damage analyses, increased chromosome aberrations (upper panel) and increased
micronuclei in cells that have reached one division (bottom panel) (n = 3 independent experiments, p < 0.05)
active in MMC treated FA cells, thus allowing their eventual division, we evaluated some
molecular markers relevant for CHKREC and cell division. We analyzed by qRT-PCR the
expression of the G2 transcriptional program, whose protein products are necessary for
the G2/M transition; namely, Cyclin A2 (CCNA2, NM_001237.3 ), Cyclin B1 (CCNB1,
NM_031966.3), WIP1 (PPM1D, NM_003620.3), FOXM1 (NM_001243088.1) and PLK1
(NM_005030.4) [16, 27, 70]. Our results show that the expression levels of these genes
remain unaffected in FA-A cells, compared to wild type cells (Fig. 5a–e). Importantly,
these genes are expressed in a cell cycle-dependent manner and are necessary for G2
phase completion [43], thus if they remain unchanged after MMC treatment, suggests
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 13 of 22
A
D
B
C
E
Fig. 5 FA cells have a gene expression pattern compatible with checkpoint resolution despite DNA damage.
Gene expression analysis of the genes belonging to the G2 transcriptional program did not show differences
in the expression of these genes despite MMC treatment. Cyclin A2 (a), Cyclin B1 (b), WIP1 (c), FOXM1 (d) and
PLK1 (e) (n = 3 independent experiments). No statistically significant differences were found in the gene
expression patterns among groups
that this program remains poised for resolution of the G2 cell cycle blockage in FA cells,
even with incomplete DNA repair.
Our BNM and these experimental results indicate that FA-A cells are able to block the
cell cycle progression in G2 but eventually recover. Simulations with the BNM showed
also that co-activation of G2 checkpoint and CHKREC components might occur in
arrested FA cells, therefore, additional to CHK1, we evaluated other protein markers to
asses key G2 checkpoint and CHKREC activation 24 h after MMC treatment. The G2
checkpoint markers included: WEE1, MYT1, p21 and γ H2AX; while CHKREC activation
markers consisted of: WIP1, Aurora A, PLK1 and CDC25B.
We observed that CHK1 activity is increased after MMC treatment in FA-A cells
(See Fig. 3b), but other checkpoint components, namely WEE1, p21 and γ H2AX (Fig. 6a),
have reduced levels. Remarkably, we also observed concomitant activation of CHKREC
proteins PLK1, CDC25B and Aurora A (Fig. 6b) in damaged FA cells. Thus indicating
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 14 of 22
A
B
Fig. 6 FA and normal cells co-express checkpoint and CHKREC proteins. a Western Blot analysis of
checkpoint proteins. b Western blot analysis of CHKREC proteins. FA cells increase the amount of some G2
blockage proteins, but have a reduction in others. Although CHK1 (Fig. 4b) and MYT1 show increased signal,
WEE1, γ H2AX and p21 protein appear as diminished in FA cells, this weakens the checkpoint blockage,
which is eventually overwhelmed by CHKREC signaling (n = 3 independent experiments, see also Fig.7)
that despite a strong CHK1 signal that leads to cell cycle progression blockage, FA-A cells
co-express the components that might dampen the DNA damage signaling and allow
an eventual CHKREC despite an elevated amount of CA. In agreement, we observed in
FA-A cells reduced levels of the histone γ H2AX, a DNA damage signaler and WIP1 phos-
phatase target. Notably, we also observed weakening of p21 protein signaling, which has
also been correlated with CHKREC activation [71]. These results suggest that signaling of
DSBs might be weakened at this time-point, triggering full CHKREC activation and cell
division despite a strong CHK1 signaling and high levels of CAs (relative protein amount
for all the markers can be seen in Fig. 7).
These experimental results show that the CHKREC is being activated in FA cells car-
rying CA to a similar extent as normal undamaged cells, this CHKREC induction might
allow the escape of G2 with unrepaired DNA damage and cell division. The correlation
between the nodes of the network and the experiments performed can be seen in Table 3.
Discussion
Several methods are used to model and analyze biological systems [72–74]. These meth-
ods analyze the topology of the network or the kinetics of the system specifying the flux
of information through a continuous model or a logical model [74].
Continuous models represent the temporal dynamics of biochemical processes with
considerable detail, but are highly dependent on the values of free parameters (initial
protein concentrations and rate constants), whose estimation might be challenging as
networks get larger [73, 75]. Logical models rely on qualitative knowledge [72]. Logical
BNM are the minimal computational model necessary to obtain a meaningful idea about
the dynamics of a regulatory network and are useful when detailed enzymatic informa-
tion is missing [75, 76]. Many molecular regulatory systems show binary behaviors or
act like bistable switches [77], thus the binary or discrete representation of BNM can
adjusts to them and predict sequence patterns of proteins and gene activities with less
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 15 of 22
A
B
D
C
E
F
H
G
I
Fig. 7 Western Blot densitometry analysis. Checkpoint proteins (a–e) and CHKREC proteins (f–i). (n = 3
independent experiments). No statistically significant differences were found in the protein expression
patterns among groups
parameters than a continuous model. Although BNM have been used for modeling sev-
eral systems [32–37], they might not be appropriate if the system has continuous values
or if knowledge on the network architecture is lacking [75].
We have developed a binary BNM that recapitulates in a simple manner the response
to ICLs mediated by the FA/BRCA pathway [78]. Given that the different components of
the network might remain unchanged, up-regulated or down-regulated instead of binary,
an additional representation of the FA/BRCA network as a discrete ternary logical net-
work might be also feasible [79], however a binary BNM resulted optimal given that
our system presents gene expression showing a pattern of binary states (over-expressed,
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 16 of 22
Table 3 Correlation between experimental validations and nodes in the FA/BRCA BNM
Process
Nodes in the
Experimental markers
Validated role
References
FA/BRCA BNM
used in this study
in the BNM
DNA damage
ICL
MMC
FA cells are
[21, 25, 26]
induction
hypersensitive
to ICL inducing
agents
Upstream
FAcore
Non-evaluated
ICL recognition
[14, 16, 17]
FA/BRCA
FANCD2I
proteins
pathway
NUC1
NUC2
DNA repair
ADD
γ H2AX,
ICLs are unhooked
[13, 21]
intermediaries
DSB
CA in metaphase
by FA core-recruited
spreads
DNA-endonucleases
that generate a DSB
and an ADD
Downstream
TLS
Non-evaluated
The ADD and DSB
[14, 15, 18, 54]
FA/BRCA
FAHRR
are repaired by TLS
pathway
and FA-dependent
downstream homologous
recombination repair,
respectively. FA cells
accumulate DSBs
Alternative
HRR2
Non-evaluated
FA cells use
[49, 56]
DNA repair
NHEJ
alternative DNA
pathways
repair pathways,
mainly NHEJ
HRR2 is a
criptic repair choice
Checkpoint
ATR
Cell cycle arrest
Upon DNA damage
[27, 28, 31]
ATM
in G2, pCHK1-S341,
normal and FA
p53
p21 gene expression,
cells activate
MYT1, WEE1, p21
the G2/M checkpoint
proteins
Checkpoint
CHKREC
MPM2 mitotic index,
The checkpoint
[83, 84] and this work
recovery
cytokinesis block assay,
is inactivated by
G2/M transcriptional
CHKREC after
program, WIP1, PLK1,
DNA repair
CDC25, Aurora A
FA cells seem to have
proteins
a lower threshold for
CHKREC activation
compared to normal cells
under-expressed) or protein concentrations that can reach a saturation regime (full acti-
vation) or remain in small concentrations (inactive). In addition the change to a ternary
system would increase the possible states of the system from 32,768 to 14,348,907 states,
thus augmenting the computational work.
Our modeling of the FA/BRCA regulatory network has led to the observation that
CHKREC is a mechanism conferring stability to this system in wild type and FA cells ([38]
and this work). CHKREC is fully activated once the G2/M checkpoint has been satisfied
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 17 of 22
leading to the division of the cell [42]. CHKREC is mainly composed of phosphatases,
such as WIP1, that inactivate the G2 checkpoint and protein-kinases that release the
cell cycle blockage, such as Aurora A and PLK1 [41, 80]. Notably, the negative circuits
mediated by CHKREC seem to be a central part of the control system of the FA/BRCA
network: they are activated when the system induces the expression of its own inhibitors,
and are necessary to attenuate the stimulatory signals arising from DNA damage (Fig. 1
and Fig. 2).
When simulating mutants, we noticed that CHKREC function inactivates the check-
point in FA core mutants despite unrepaired DNA damage, thus resolving the
G2/M checkpoint arrest and allowing cell division. Therefore we should notice ele-
vated/unchanged levels in the expression, quantity or activity of CHKREC components
in FA cells with damaged DNA compared to undamaged normal cells, indicating
that FA cells conserve checkpoint resolution capacity and are poised for cell divi-
sion when the DNA damage checkpoint response ceases. To test the function of the
CHKREC node, we experimentally evaluated the cell division capacity as well as check-
point/CHKREC activation in FA-A lymphoblasts after induction of DNA damage with
MMC.
We evaluated the G2 blockage and found accumulation of FA-A cells into the G2 phase
compartment after induction of DNA damage (Fig. 3a left panel) and a reduced num-
ber of FA-A mitotic cells in comparison to normal cells after MMC exposition (Fig. 3a
right panel). We also detected high CHK1 phosphorylation levels (Fig. 3b) as well as high
p21 gene expression (Fig. 3c) in FA-A cells. CHK1 is a key protein kinase that transduces
the DNA damage signaling, and p21 is a direct p53 transcription target, therefore an
increase in p21 activation is the result of p53-increased activity, thus demonstrating that
FA cells achieve a correct activation of the checkpoint that blocks the G2/M transition
[27, 28]. p21 is a negative regulator of Cyclin B/CDK1 complex and is necessary to avoid
the G2/M transition in presence of DNA damage [81]. Thus, CHK1 phosphorylation and
p21 expression augment when a cell is exposed to DNA damaging agents and would be
expected to drop-off once a cell has repaired the DNA damage [82].
When we evaluated the cell division capacity in a CB block assay, we did not observe
differences in the frequency of binucleated cells between normal and FA-A cells (Fig. 4
middle panel), although MMC limited tetranucleated cells production in both cell types
(Fig. 4c bottom panel). These results show that, under these experimental conditions,
both FA-A and normal cells divide to a similar extent after induction of DNA damage by
MMC.
The capability of FA cells to divide with unrepaired DNA damage was evaluated by
quantifying the frequency of DNA damage induced by MMC in cells committed to divide
by scoring CAs in metaphase spreads, as well as in cells that have already performed cell
division by quantifying the micronuclei observed in binucleated and tetranucleated cells.
Our results showed, in both assays, that FA-A cells exposed to MMC carry significant
DNA damage during mitosis and, nonetheless divide (Fig. 4b).
Our BNM allowed us to propose that CHKREC function in FA cells might ignore
in a certain level the presence of unrepaired DNA damage and could be responsible
for their division, therefore we expected that normal and FA-A cells would have simi-
lar activation levels of the CHKREC components. To test this possibility, we measured
the expression of the G2 transcriptional program genes that promote CDK activity and
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 18 of 22
progression into mitosis, namely WIP1, Cyclin A2, Cyclin B1, PLK1, CDC25 and FOXM1
[43] (Fig. 5a–e) and evaluated the activation of some of the proteins involved in check-
point and CHKREC (Fig. 6). In the first assay we observed that the expression of the genes
that enable CHKREC and cell division are similar in normal/undamaged cells and dam-
aged FA cells, even when FA cells carry a higher number of CAs. In addition, we observed
co-expression of checkpoint and CHKREC proteins in FA cells treated with MMC (Fig. 6
and 7), indicating that damaged FA cells are poised for an eventual cell division despite
DNA damage (Fig. 8).
Checkpoint activation, cell cycle arrest and DNA repair require a great number of
protein posttranslational modifications for their establishment. Dedicated enzymes that
remove these modifications or degrade modified proteins allow checkpoint silencing and
recovery [84]. WIP1 phosphatase and PLK1 kinase emerge as the coordinators of check-
point silencing and recovery, respectively, however if there exist a certain order in their
activation remains elusive. In general terms for cell division, Cyclin B levels must gradu-
ally increase, while CDC25 phosphatase should remove any inhibitory phosphorylation of
CDK1, thus promoting Cyclin B/CDK1 complex formation and mitotic entry. However,
after induction of DNA damage, the G2 checkpoint inhibits CDK1 activity through p21,
whilst WEE1 and MYT1 kinases degrade CDC25, avoiding mitotic entry [42, 70, 80].
WIP1 phosphatase dephosphorylates ATM, p53, CHK1, CHK2, γ H2AX and the
p(S/T)Q motif originally modified by ATM and ATR [85, 86]. During CHKREC, Aurora-
A kinase activates PLK1, which in turn targets WEE1 for proteasomal degradation and
releases CDK1 from blockage [42, 87–89], in addition PLK1 interferes with CHK1,
CHK2 and p53 stability, thus it also has an active role turning-off the DNA damage
checkpoint [40].
Fig. 8 CHKREC components activation in normal and FA cells. After DNA damage induction, the cell activates
the DNA damage integrity checkpoints, the G2/M checkpoint specifically avoids the transition of the cell
from G2 to M phase with unrepaired DNA damage. Once the DNA has been repaired, the G2/M checkpoint is
satisfied and the cell activates the CHKREC, a process that inactivates checkpoint proteins and promotes cell
division. Upper panel. Normal cells activate CHKREC with repaired chromosomes. Bottom panel. FA cells
activate CHKREC despite unrepaired chromosomes. The specific mechanism triggering this inappropriate
CHKREC activation in FA cells remains unknown
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 19 of 22
In Fig. 6b we observe that the concentration of WIP1 is increased in normal cells and
reduced in FA cells; on the contrary PLK1 is reduced in normal cell and increased in
FA cells. Interestingly, PLK1 activity is redundant in unperturbed mitotic entry whereas
it becomes essential in CHKREC after DNA damage [88, 89], consistently it is activated
in our experiments in damaged FA-A cells. As FA cells carry spontaneous unrepaired
DNA damage, this implies that their transition through G2 is always perturbed to a
certain extent, thus PLK1 should become essential for FA cells survival. Given this,
PLK1 over-activation must be involved in the adaptation of FA cells to DNA dam-
age. Recent evidence shows that PLK1 activity is gradually increased during an ongoing
DNA damage-induced cell cycle arrest and if the activity of the kinase exceeds beyond
a certain level, the cell progresses to mitosis despite DNA damage persistance [83]. G2
checkpoint recovery might thus represent a checkpoint adaptation, where DNA dam-
age triggers an arrest whose duration is not necessarily conditioned by DNA repair [84].
Regarding this, PLK1 might have a more critical role than WIP1 in the delivery of FA
cells with unrepaired DNA damage from the G2 arrest, or WIP1 is acting before than
the time-point that we are evaluating in this assays, hence we are not able to detect
WIP1 protein (Fig. 6b). The distinction between both possible scenarios deserves further
research.
A final aspect to be considered are the findings of Ceccaldi and coworkers [31], who
described an attenuated G2/M checkpoint activity in adult FA individuals that, con-
comitantly to low CHK1 and p53 protein levels, allowed the escape of unrepaired DNA
damage. Although they demonstrate that downregulation of the ATR-CHK1 axis is
responsible for this phenotype, it remains elusive if this reduced checkpoint activity might
be due to CHKREC over-activation or ectopic activity. In this study we set the basis to
explore this possibility in FA individuals with an attenuated G2/M checkpoint and the
general mechanism allowing G2/M resolution in non-attenuated FA individuals. Further,
modeling the full interaction between the G2/M checkpoint and CHKREC, as well as a
systematic inhibition of CHKREC components in FA cells, will shed light into the intricate
interactions between these two processes.
Our results show that highly damaged FA-A cells preserve the capacity to divide after a
cell cycle arrest induced by DNA damage, a result that is consistent with our BNM FA core
null mutant simulations. Nonetheless, the definition of the specific trigger for cell division
remains unknown. To our judgment, the CHKREC hypothesis became the most rele-
vant hypothesis emerging from our BNM given that CHKREC promotion might enable
cell survival and amelioration of blood cell counts in pancytopenic FA patients, however
CHKREC overexpression might also lead to exhaustion of the hematopoietic stem cell
compartment as well as selection of malignant clones. Therefore, the thorough study of
this process becomes relevant for the understanding of hematopoiesis and carcinogenesis
in a FA background.
Conclusion
In this study we propose through network modeling that CHKREC, a program neces-
sary for cell division after DNA damage, becomes activated in FA core mutant cells with
unrepaired DNA damage. We experimentally show that highly damaged FA-A cells have
CHKREC expression levels similar to those observed in normal undamaged cells, thus
FA-A cells might ignore the presence of broken chromosomes through this process. We
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 20 of 22
observed that despite a prominent G2 arrest after MMC exposure, FA cells were able to
activate the mechanisms that allow cell division (Fig. 8).
FA cells are prone to apoptosis due to their DNA repair defects, however a great quan-
tity of them divide in spite of unrepaired DNA damage, thus allowing the survival of
FA individuals. The study of the mechanisms that allow FA cells to survive may help
to develop novel therapies designed to promote hematopoiesis, as well as to avoid the
division of malignant clones in FA patients.
Competing interests
The authors declare that they have no competing interests.
Authors’ contributions
AR, LM and SF conceived the project; AR, DS and EA developed the BNM; AR, LT, UJ, DS, BGT, EC and AMS performed
experiments; RO and POW provided essential reagents; LM coordinated computational work; SF coordinated laboratory
work. All authors read and approved the final manuscript.
Acknowledgements
Authors thank our colleagues at Dr. Frías’ laboratory for insightful discussions and Anet Rivera Osorio for invaluable
assistance with experiments. This work was supported in part by grants from Universidad Nacional Autónoma de México
PAPIIT IN200514 and IA201713, and Instituto Nacional de Pediatría-SSA, Fondos Federales 043-12. AR received the
346717 scholarship from Consejo Nacional de Ciencia y Tecnología (CONACyT). We thank S Becerra and J Yañez from IBT
UNAM for primer synthesis.
Author details
1Laboratorio de Citogenética, Departamento de Investigación en Genética Humana, Instituto Nacional de Pediatría, D.F.,
México. 2Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, D.F., México.
3Instituto de Ecología, Universidad Nacional Autónoma de México, D.F., México. 4C3, Centro de Ciencias de la
Complejidad, Universidad Nacional Autónoma de México, D.F., México. 5Current address: INRIA, Virtual Plants Project
Team, UMR AGAP, Montpellier, France. 6Departamento de Ciencias de la Salud, Universidad Autónoma
Metropolitana-Iztapalapa, D.F., México. 7Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de
México, D.F., México.
Received: 26 May 2015 Accepted: 12 August 2015
References
1.
Surralles J, Jackson SP, Jasin M, Kastan MB, West SC, Joenje H. Molecular cross-talk among chromosome fragility
syndromes. Genes Dev. 2004;18(12):1359–70.
2.
Aurias A, Antoine JL, Assathiany R, Odievre M, Dutrillaux B. Radiation sensitivity of bloom’s syndrome lymphocytes
during S and G 2 phases. Cancer Genet Cytogenet. 1985;16(2):131–6.
3.
Krepinsky AB, Heddle JA, German J. Sensitivity of Bloom’s syndrome lymphocytes to ethyl methanesulfonate. Hum
Genet. 1979;50(2):151–6.
4.
Taylor AMR. Unrepaired DNA strand breaks in irradiated ataxia telangiectasia lymphocytes suggested from
cytogenetic observations. Mutat Res Fundam Mol Mech Mutagen. 1978;50(3):407–18.
5.
German J, Reginald A, David B. Chromosomal breakage in a rare and probably genetically determined syndrome of
man. Science. 1965;148.3669:506–7.
6.
German J, Sanz MM, Ciocci S, Ye TZ, Ellis NA. Syndrome causing mutations of the BLM gene in persons in the
Bloom’s Syndrome Registry. Hum Mutat. 2007;28(8):743–53.
7.
Chaganti RSK, Schonberg S, German J. A manyfold increase in sister chromatid exchanges in Bloom’s syndrome
lymphocytes. Proc Natl Acad Sci. 1974;71(11):4508–12.
8.
Tadjoedin MK, Fraser FC. Heredity of ataxia-telangiectasia (Louis-Bar syndrome). Am J Dis Child. 1965;110(1):64–8.
9.
Concannon P, Gatti RA. Diversity of ATM gene mutations detected in patients with ataxia-telangiectasia. Hum
Mutat. 1997;10(2):100.
10. Kojis TL, Gatti RA Sparkes, RS. The cytogenetics of ataxia telangiectasia. Cancer Genet Cytogenets. 1991;56(2):
143–156.
11. Oxford JM, Harnden DG, Parrington JM, Delhanty JD. Specific chromosome aberrations in ataxia telangiectasia.
J Med Genet. 1975;12(3):251–62.
12. Lobitz S, Velleuer E. Guido Fanconi (1892–1979): a jack of all trades. Nat Rev Cancer. 2006;6(11):893–8.
13. de Winter J, Joenje H. The genetic and molecular basis of Fanconi anemia. Mutat Res. 2009;668:11–9.
14. Kee Y, D’Andrea AD. Expanded roles of the Fanconi anemia pathway in preserving genomic stability. Genes Dev.
2010;24:1680–94.
15. Vaz F, Hanenberg H, Schuster B. Mutation of the RAD51C gene in a fanconi anemia-like disorder. Nat Genet.
2010;42:406–9.
16. Kim Y, Lach F, Desetty R, et al. Mutations of the SLX4 gene in fanconi anemia. Nat Genet. 2011;43:142–6.
17. Bogliolo M, Schuster B, Stoepker C, et al. Mutations in ERCC4, encoding the DNA-repair endonuclease XPF cause
Fanconi anemia. Am J Hum Genet. 2013;92:800–6.
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 21 of 22
18. Greenberg RA, Sawyer SL, Tian L, et al. Biallelic mutations in BRCA1 cause a new Fanconi anemia subtype. Cancer
discov.2014;CD-14.
19. Hira A, Yoshida K, Sato K, Okuno Y, Shiraishi Y, Chiba K, et al. Mutations in the Gene Encoding the E2 Conjugating
Enzyme UBE2T Cause Fanconi Anemia. Am J Hum Genet. 2015;96(6):1001–7.
20. Schroeder TM, German J. Bloom’s syndrome and Fanconi’s anemia: demonstration of two distinctive patterns of
chromosome disruption and rearrangement. Humangenetik. 1974;25(4):299–306.
21. Auerbach A. Fanconi anemia and its diagnosis. Mutat Res. 2009;668:4–10.
22. Amor-Guéret M. Bloom syndrome, genomic instability and cancer: the SOS-like hypothesis. Cancer Lett. 2006;236(1):
1–12.
23. Soulier J. Fanconi anemia. Hematol Am Soc Hematol Educ Prog. 2011492–7.
24. Tulpule A, Lensch MW, Miller J, et al. Knockdown of Fanconi anemia genes in human embryonic stem cells reveals
early developmental defects in the hematopoietic lineage. Blood. 2011;115(17):3453–62.
25. Langevin F, Crossan GP, Rosado IV, et al. Fancd2 counteracts the toxic effects of naturally produced aldehydes in
mice. Nature. 2011;475:53–8.
26. Rosado IV, Langevin F, Crossan GP, et al. Formaldehyde catabolism is essential in cells deficient for the Fanconi
anemia DNA-repair pathway. Nat Struct Mol Biol. 2011;18:1432–4.
27. Ceccaldi R, Parmar K, Mouly E, et al. Bone marrow failure in Fanconi anemia is triggered by an exacerbated p53/p21
DNA damage response that impairs hematopoietic stem and progenitor cells. Cell Stem Cell. 2012;11:36–49.
28. Heinrich M, Hoatlin ME, Zigler AJ, et al. DNA cross-linker-induced G2/M arrest in group C Fanconi anemia
lymphoblasts reflects normal checkpoint function. Blood. 1998;91:275–87.
29. Freie B, Ciccone S, Li X, et al. A role for the Fanconi anemia C protein in maintaining the DNA damage-induced G2
checkpoint. J Biol Chem. 2004;279:50986–93.
30. Neveling K, Endt D, Hoehn H, et al. Genotype-phenotype correlations in Fanconi anemia. Mutat Res. 2009;668:73–91.
31. Ceccaldi R, Briot D, Larghero J, et al. Spontaneous abrogation of the G2 DNA damage checkpoint has clinical
benefits but promotes leukemogenesis in Fanconi anemia patients. J Clin Invest. 2011;121:184–94.
32. Faure A, Naldi A, Chaouiya C, et al. Dynamical analysis of a generic boolean model for the control of the
mammalian cell cycle. Bioinformatics. 2006;22:e124–131.
33. Mendoza L. A network model for the control of the differentiation process in Th cells. Biosystems. 2006;84:101–14.
34. Zhang R, Shah MV, Yang J, et al. Network model of survival signaling in large granular lymphocyte leukemia. Proc
Natl Acad Sci USA. 2009;105:16308–13.
35. Chaves M, Albert R, Sontag E. Robustness and fragility of Boolean models for genetic regulatory networks. J Theor
Biol. 2005;235:431–49.
36. Christensen C, Thakar J, Albert R. Systems-level insights into cellular regulation: inferring, analyzing, and modelling
intracellular networks. IET Syst Biol. 2007;1:61–77.
37. Wu M, Yang X, Chan C. A dynamic analysis of IRS-PKR signaling in liver cells: a discrete modeling approach. PLoS
One. 2009;4(12):e8040.
38. Rodriguez A, Sosa D, Torres L, et al. A Boolean network model of the FA/BRCA pathway. Bioinformatics. 2012;28:
858–66.
39. van Vugt M, Yaffe M, et al. Cell cycle re-entry mechanisms after DNA damage checkpoints: Giving it some gas to
shut off the breaks. Cell Cycle. 2010;11:2097–101.
40. van Vugt MA, Gardino AK, Linding R, et al. A mitotic phosphorylation feedback network connects Cdk1, Plk1, 53BP1
and Chk2 to inactivate the G2/M DNA damage checkpoint. PLoS Biol. 2010;8:e1000287.
41. Halim V, Alvarez-Fernandez M, Xu YJ, et al. omparative phosphoproteomic analysis of checkpoint recovery
identifies new regulators of the DNA damage response. Sci Signal. 2013;6:rs9.
42. Medema RH, Macurek L. Checkpoint recovery in cells: how a molecular understanding can help in the fight against
cancer. F1000 Biol Rep. 2011;3.
43. Álvarez-Fernández M, Medema RH, Lindqvist A. Transcriptional regulation underlying recovery from a DNA
damage-induced arrest. Transcription. 2010;1:32–5.
44. Meetei AR, et al. A Human Orthologue of Archaeal DNA Repair Protein Hef is Defective in Fanconi Anemia
Complementation Group. M Nat Genet. 2005;37:958–63.
45. Kee Y, et al. Regulated degradation of FANCM in the Fanconi anemia pathway during mitosis. Genes Dev. 2009;23:
555–60.
46. Wang XZ, et al. Chk1-Mediated Phosphorylation of FANCE Is Required for the Fanconi Anemia/BRCA Pathway. Mol
Cell Biol. 2007;27:3098–108.
47. Smogorzewska A, et al. Identification of the Fanconi anemia (FANC) I protein, a monoubiquitinated FANCD2 paralog
required for crosslink repair. Cell. 2007;129:289–301.
48. Bogliolo M, et al. Histone H2AX and Fanconi anemia FANCD2 function in the same pathway to maintain
chromosome stability. EMBO J. 2007;26:1340–51.
49. Pace P, et al. Ku70 Corrupts DNA Repair in the Absence of the Fanconi Anemia Pathway. Science. 2010;329:219–23.
50. Bhagwat N, et al. XPF-ERCC1 Participates in the Fanconi Anemia Pathway of Cross-Link Repair. Mol Cell Biol. 2009;29:
6427–37.
51. Hanada K, et al. The structure-specific endonuclease Mus81/Eme1 promotes conversion of interstrand DNA
crosslinks into double-strands breaks. EMBO J. 2006;25:492125–4932.
52. Lieber MR. The Mechanism of Double-Strand DNA Break Repair by the Nonhomologous DNA End-Joining Pathway.
Annu Rev Biochem. 2010;79:181–211.
53. Chang DJ, Cimprich KA. DNA damage tolerance: when it’s OK to make mistakes. Nature Chem Biol. 2009;5:82–90.
54. Mirchandani KD, et al. The Fanconi anemia core complex is required for efficient point mutagenesis and Rev1 foci
assembly. DNA Repair. 2008;7:902–11.
55. Kass EM, Jasin M. Collaboration and competition between DNA double-strand break repair pathways. FEBS Letters.
2010;584:3703–8.
Rodríguez et al. Theoretical Biology and Medical Modelling (2015) 12:19
Page 22 of 22
56. Adamo A, et al. Preventing Non-homologous End-Joining suppresses DNA repair defects of Fanconi Anemia. Mol
Cell. 2010;39:25–35.
57. Yaneva M, et al. Interaction of DNA-dependent protein kinase with DNA and with KU: biochemical and atomic-force
microscopy studies. EMBO J. 1997;16:5098–112.
58. Cimprich KA, Cortez D. ATR: an essential regulator of genome integrity. Nat Rev Mol Cell Biol. 2008;9:616–127.
59. Collis SJ, et al. FANCM and FAAP24 function in ATR- Mediated Checkpoint Signaling Independently of the Fanconi
Anemia Core Complex. Mol Cel. 2008;32:313–24.
60. Jazayeri A, et al. ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks. Nature
Cell Biol. 2006;8:37–45.
61. Stiff T, et al. ATR-dependent phosphorylation and activation of ATM in response to UV treatment or replication fork
stalling. EMBO J. 2006;25:5775–82.
62. Lavin MF. ATM and the Mre11 complex combine to recognize and signal DNA double-strand breaks. Oncogene.
2008;26:7749–58.
63. Lee J, et al. The Rad9-Hus1-Rad1 Checkpoint Clamp regulates interaction of TopBP1 with ATR. J Biol Chem. 2007;282:
28036–44.
64. Shieh S, et al. The human homologs of checkpoint kinases Chk1 and Cds1 (Chk2) phosphorylate p53 at multiple
DNA damage-inducible sites. Genes Dev. 2000;14:289–300.
65. Bassermann F, Pagano M. Dissecting the role of ubiquitylation in the DNA damage response checkpoint in G2. Cell
Death Differ. 2010;17:78–85.
66. Mussel C, Hopfensitz M, Kestler HA. BoolNet-an R package for generation, reconstruction and analysis of Boolean
networks. Bioinformatics. 2010;26:1378–80.
67. Fenech M, Morley AA. Measurement of micronuclei in lymphocytes. Mutat Res. 1985;147:29–36.
68. Zhou B, Elledge SJ. The DNA damage response: putting checkpoints in perspective. Nature. 2000;408:433–9.
69. Khanna KK, Jackson SP. DNA double-strand breaks: signaling, repair and the cancer connection. Nat Genet. 2001;27:
247–54.
70. Bloom J, Cross FR. Multiple levels of cyclin specificity in cell-cycle control. Nat Rev Mol Cell Biol. 2007;8:149–60.
71. Krenning L, Feringa FM, Shaltiel IA, et al. Transient activation of p53 in G2 phase is sufficient to induce senescence.
Molecular Cell. 2014;55:59–72.
72. Karlebach G, Shamir R. Modelling and analysis of gene regulatory networks. Nat Rev Mol Cell Biol. 2008;9(10):770–80.
73. Morris MK, Saez-Rodriguez J, Sorger PK, Lauffenburger DA. Logic-based models for the analysis of cell signaling
networks. Biochemistry. 2010;49(15):3216–24.
74. Tenazinha N, Vinga S. A survey on methods for modeling and analyzing integrated biological networks. IEEE/ACM
Trans Comput Biol Bioinforma (TCBB). 2011;8(4):943–58.
75. Bornholdt S. Boolean network models of cellular regulation: prospects and limitations. J R Soc Interface 5. 2008;1:
S85–S94.
76. Lazebnik Y. Can a biologist fix a radio?—or, what I learned while studying apoptosis. Biochemistry (Moscow).
2004;69(12):1403–6.
77. Tyson JJ, Chen KC, Novak B. Sniffers, buzzers, toggles and blinkers: dynamics of regulatory and signaling pathways
in the cell. Curr Opin Cell Biol. 2003;15(2):221–31.
78. Wang AT, Smogorzewska A. SnapShot: Fanconi anemia and associated proteins. Cell. 2015;160(1):354.
79. Fritsch P, Craddock TJA, del Rosario RM, Rice MA, Smylie A, Folcik VA, et al. Succumbing to the laws of attraction:
Exploring the sometimes pathogenic versatility of discrete immune logic. Syst Biomed. 2013;1(3):179–94.
80. Peng A. Working hard for recovery: mitotic kinases in the DNA damage checkpoint. Cell Biosci. 2013;3:20.
81. Amador V, Ge S, Santamaría PG, et al. APCCDC20controls the ubiquitin-mediated degradation of p21 in
prometaphase. Mol Cell. 2007;3:462–73.
82. Mailand N, Bekker-Jensen S, Bartek J, et al. Destruction of Claspin by SCFbTrCP restrains Chk1 activation and
facilitates recovery from genotoxic stress. Mol Cell. 2006;23:307–18.
83. Liang H, Esposito A, De S, Ber S, Collin P, Surana U, et al. Homeostatic control of polo-like kinase-1 engenders
non-genetic heterogeneity in G2 checkpoint fidelity and timing. Nat Commun. 2014;5.
84. Shaltiel IA, Krenning L, Bruinsma W, Medema RH. The same, only different—DNA damage checkpoints and their
reversal throughout the cell cycle. J Cell Sci. 2015;128:607–20.
85. Lindqvist A, de Bruijn M, Macurek L, et al. Wip1 confers G2 checkpoint recovery competence by counteracting p53
dependent transcriptional repression. EMBO J. 2009;20:3196–206.
86. Shaltiel IA, Aprelia M, Saurin AT, et al. Distinct phosphatases antagonize the p53 response in different phases of the
cell cycle. Proc Natl Acad Sci USA. 2014;20:7313–8.
87. Seki A, Coppinger J, Jang C, et al. Bora and the kinase Aurora A cooperatively activate the kinase Plk1 and control
mitotic entry. Science. 2008;320:1655–8.
88. Macurek L, Lindqvist A, Lim D, et al. Polo-like kinase-1 is activated by aurora A to promote checkpoint recovery.
Nature. 2008;455:119–23.
89. van Vugt MA, Brás A, Medema RH. Polo-like kinase-1 controls recovery from a G2 DNA damage-induced arrest in
mammalian cells. Mol Cell. 2004;15(5):799–811.
|
26385365
|
ATR = ( ( ATM ) AND NOT ( CHKREC ) ) OR ( ( ICL ) AND NOT ( CHKREC ) )
FAHRR = ( ( FANCD2I AND ( ( ( DSB ) ) ) ) AND NOT ( NHEJ AND ( ( ( CHKREC ) ) ) ) )
ICL = ( ( ICL ) AND NOT ( DSB ) )
p53 = ( ( ATM ) AND NOT ( CHKREC ) ) OR ( ( NHEJ ) AND NOT ( CHKREC ) ) OR ( ( ATR ) AND NOT ( CHKREC ) )
ATM = ( ( ( DSB ) AND NOT ( CHKREC ) ) AND NOT ( FAcore ) ) OR ( ( ( ATR ) AND NOT ( CHKREC ) ) AND NOT ( FAcore ) )
HRR2 = ( ( ( ( TLS AND ( ( ( NUC2 AND DSB ) ) ) ) AND NOT ( FAHRR ) ) AND NOT ( CHKREC ) ) AND NOT ( NHEJ ) ) OR ( ( ( NUC2 AND ( ( ( DSB AND ICL AND NHEJ ) ) ) ) AND NOT ( FAHRR ) ) AND NOT ( CHKREC ) )
NUC1 = ( FANCD2I AND ( ( ( ICL ) ) ) )
ADD = ( ( ( NUC1 ) AND NOT ( TLS ) ) AND NOT ( TLS ) ) OR ( ( NUC2 ) AND NOT ( TLS ) )
CHKREC = ( ( ( FAHRR ) AND NOT ( DSB ) ) OR ( ( TLS ) AND NOT ( DSB ) ) OR ( ( HRR2 ) AND NOT ( DSB ) ) OR ( ( NHEJ ) AND NOT ( DSB ) ) ) OR NOT ( TLS OR FAHRR OR DSB OR ADD OR HRR2 OR ICL OR CHKREC OR NHEJ )
DSB = ( ( ( ( NUC2 ) AND NOT ( FAHRR ) ) AND NOT ( HRR2 ) ) AND NOT ( NHEJ ) ) OR ( ( ( ( NUC1 ) AND NOT ( FAHRR ) ) AND NOT ( HRR2 ) ) AND NOT ( NHEJ ) )
TLS = ( ( FAcore AND ( ( ( ADD ) ) ) ) AND NOT ( CHKREC ) ) OR ( ( ADD ) AND NOT ( CHKREC ) )
FANCD2I = ( ( FAcore AND ( ( ( DSB ) AND ( ( ( ATR OR ATM ) ) ) ) OR ( ( ATR OR ATM ) ) ) ) AND NOT ( CHKREC ) )
FAcore = ( ( ICL AND ( ( ( ATR OR ATM ) ) ) ) AND NOT ( CHKREC ) )
NUC2 = ( ( ICL AND ( ( ( ATR OR ATM ) ) ) ) AND NOT ( FAcore AND ( ( ( FANCD2I ) ) ) ) ) OR ( ( NUC1 AND ( ( ( ICL AND p53 ) ) ) ) AND NOT ( FAcore AND ( ( ( FANCD2I ) ) ) ) )
NHEJ = ( ( ( ( NUC2 AND ( ( ( DSB ) ) ) ) AND NOT ( HRR2 ) ) AND NOT ( CHKREC ) ) AND NOT ( FAHRR ) )
|
1
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
www.nature.com/scientificreports
Network modelling reveals the
mechanism underlying colitis-
associated colon cancer and
identifies novel combinatorial
anti-cancer targets
Junyan Lu1,*, Hanlin Zeng1,*, Zhongjie Liang2,*, Limin Chen1, Liyi Zhang1, Hao Zhang1,
Hong Liu1, Hualiang Jiang1, Bairong Shen2, Ming Huang1, Meiyu Geng1, Sarah Spiegel3 &
Cheng Luo1,2
The connection between inflammation and tumourigenesis has been well established. However, the
detailed molecular mechanism underlying inflammation-associated tumourigenesis remains unknown
because this process involves a complex interplay between immune microenvironments and epithelial
cells. To obtain a more systematic understanding of inflammation-associated tumourigenesis as
well as to identify novel therapeutic approaches, we constructed a knowledge-based network
describing the development of colitis-associated colon cancer (CAC) by integrating the extracellular
microenvironment and intracellular signalling pathways. Dynamic simulations of the CAC network
revealed a core network module, including P53, MDM2, and AKT, that may govern the malignant
transformation of colon epithelial cells in a pro-tumor inflammatory microenvironment. Furthermore,
in silico mutation studies and experimental validations led to a novel finding that concurrently
targeting ceramide and PI3K/AKT pathway by chemical probes or marketed drugs achieves
synergistic anti-cancer effects. Overall, our network model can guide further mechanistic studies on
CAC and provide new insights into the design of combinatorial cancer therapies in a rational manner.
Inflammation and cancer are closely correlated1. The link between inflammation and cancer development
is especially strong in patients with colorectal cancer (CRC), which is one of the most common malig-
nancies and a leading cause of cancer mortality worldwide2. An increased risk of CRC development has
been observed in patients with inflammatory bowel disease (IBD)3, and nonsteroidal anti-inflammatory
drugs are effective in preventing colon neoplasia4. Dysregulations of the immune microenvironment and
several inflammation-related signalling pathways, such as TNF-α /NF-κ B, IL-6/STAT3, COX-2/PGE2 and
TGF-β /SMADs, have been shown to contribute to the development of inflammation-associated cancers5–9.
In addition, emerging evidence suggests a possible link between the inflammatory microenvironment
and cancer therapy resistance10. Nevertheless, most of these studies have focused on a single molecule
or pathway. Information on how the immune microenvironment affects cancer development and how
the inflammatory signalling pathways crosstalk with classical tumourigenesis pathways is still lacking.
Therefore, to gain a holistic view on the mechanism of the development of inflammation-associated
1State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences,
Shanghai, China. 2Soochow University, Center for Systems Biology, Jiangsu, China. 3Department of Biochemistry
and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA. *These
authors contributed equally to this work. Correspondence and requests for materials should be addressed to
M.H. (email: mhuang@simm.ac.cn) or M.G. (email: mygeng@simm.ac.cn) or C.L. (email: cluo@simm.ac.cn)
received: 15 January 2015
accepted: 07 September 2015
Published: 08 October 2015
OPEN
www.nature.com/scientificreports/
2
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
cancers, as well as to identify effective therapeutic targets, the extracellular microenvironment and intra-
cellular signalling should be considered as a complex system and studied in a more systematic manner.
To date, network modelling has been successfully used in the study of complex biological systems11–13.
Existing knowledge of individual pathways can be incorporated into an integrated biological network,
which could be further converted into a dynamic and predictive model using various mathematical
modelling techniques. Boolean network models are the simplest discrete mathematical models and
assume only two states (ON or OFF) for each node in the biological networks. Dynamic Boolean net-
work models have been successfully applied in studies of complex diseases and biological processes,
such as survival signalling of T-cell large granular lymphocyte (T-LGL) leukaemia13, hepatocyte growth
factor (HGF)-induced keratinocyte migration12, immune cell differentiation14, and cell cycle regulation11.
Boolean network models have also been used to integrate microenvironment components and signaling
pathways to study cancer biology and predict therapy outcomes15,16. Boolean network models are espe-
cially useful when the biochemical kinetic parameters of a certain biological process are unknown or
the networks contain different species of biological entities, such as proteins, small molecules, mRNAs,
and even cells.
In the present work, we constructed a Boolean network model describing the growth and survival
of preneoplastic epithelial cells in an inflammatory microenvironment, aiming to systematically study
the molecular mechanisms underlying the development of colitis-associated colon cancer (CAC). The
ability of the network model to recapture experimental observations validated its rationality. The detailed
dynamic properties of the CAC network model under normal or dysregulated inflammatory microen-
vironments were characterised. Our simulation results suggest the constant activation of the node rep-
resenting dendritic cells (DC) creates a pro-tumor inflammatory microenvironment. Attractor analysis
identified a key regulatory module involving P53, MDM2, GSK3-β and AKT signalling that may govern
the malignant transformation of epithelial cells in this pro-tumour inflammatory microenvironment.
Furthermore, in silico perturbation studies and experimental validations led us to identify several novel
drug combinations that could significantly inhibit proliferation and induce apoptosis of tumour cells
under an inflammatory stimulus. Taken together, our study integrates the extracellular microenviron-
ment and intracellular signalling to provide a holistic view of inflammation-associated cancer. Our dry
lab model and experimental findings can accelerate mechanistic studies and the development of novel
combinatorial therapies for CAC and other inflammation-associated cancers.
Results
The CAC network representing intestinal epithelial cells in an immune microenvironment.
By
performing extensive literature and database searches, we constructed a knowledge-based network link-
ing inflammatory signalling and cell proliferation and survival pathways of premalignant intestinal epi-
thelial cells (IECs) (Fig. 1). We designated this network model as the CAC network. The entire CAC
network incorporates 70 nodes and 153 edges. It can be divided into two parts: the IEC part, which
contains nodes representing intracellular signalling components, and the immune microenvironment
part, which contains the nodes representing immune cells, cytokines and chemokines. We also mod-
elled ‘Proliferation’ and ‘Apoptosis’ as two output nodes to summarise the final biological effects of the
inflammatory signalling. The nomenclature of all the nodes in the network is provided in Supplementary
Table S1, and the biological description of the CAC network is presented in Supplementary Methods.
The topology properties of the CAC network are summarised in Supplementary Table S2. The properties
of the CAC network resemble those of general biological networks, which are characterised by higher
clustering coefficient than random networks (Supplementary Table S2) and approximate power-law dis-
tributions of node degrees (Supplementary Fig. S1). However, as the size of the CAC network is relatively
small and its topology has been simplified, the quantitative characterisation of the network topology is
not very informative and therefore we are focusing more on the dynamic properties of the CAC network.
To further characterise the dynamic cell signaling events, we translated the CAC network into a
Boolean network model, in which the network node was described by one of two possible states: ON or
OFF. The ON state can be biologically interpreted as the activation of a gene/protein, or the production
of a small molecule whereas the OFF state means the inhibition of a gene/protein or the absence of
a small molecule. The regulatory relationships between upstream nodes (regulators) and downstream
nodes (targets) are expressed by the logical operators AND, OR and NOT. The Boolean logical rules that
govern the states of all these nodes are listed in Supplementary Table S3 and a thorough justification
of these rules is provided in Supplementary Methods. Preliminary robustness tests suggested the CAC
network model was robust to small amounts of noise (Supplementary Fig. S2), which is in accord with
the general feature of biological networks.
Dynamics of the CAC network model in a normal immune microenvironment.
We first exam-
ined whether our CAC network model could reproduce the experimental observations of the IECs in
a normal immune microenvironment, including a non-inflammatory microenvironment and a normal
inflammatory response. To simulate a non-inflammatory microenvironment, we fixed the states of all
the nodes in the immune microenvironment (cyan nodes in Fig. 1) to OFF to represent the absence of
inflammatory factors. We also fixed the state of the APC node to ON to represent premalignant IECs, in
which the adenomatous polyposis coli (APC) protein is constantly expressed and activated to suppress
www.nature.com/scientificreports/
3
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
β -catenin signalling17. Subsequently, we iterated the model using the general asynchronous (GA) updat-
ing method from a large number (5,000) of randomly selected initial states. When simulating a dynamic
Boolean model using asynchronous updating methods, the frequency of a node being in the ON state
(activation frequency) can give qualitative indications of the probability that a certain signaling compo-
nent or a biological process being activated in a real cell. The activation frequencies of the two output
nodes – Proliferation and Apoptosis at each simulation step were recorded to evaluate the impact of the
microenvironment on cell survival states (Fig. 2a). In order to facilitate the comparison between our
simulation results and previous experimental observations, we also recorded the activation frequen-
cies of three nodes representing STAT3, NF-κ B and β -catenin transcription factors (STAT3, NFKB and
BCATENIN), whose activations have been considered as hallmarks of CAC17–20. After 1000 steps of
iteration, the Proliferation node rested in the OFF state, suggesting the IECs were unable to proliferate
under non-inflammatory microenvironment. STAT3, NFKB and BCATENIN also stabilised in the OFF
state within 1500 steps (Fig. 2a). By contrast, the steady activation frequency of the Apoptosis node
was approximately 30%, indicating that a fraction of the epithelial cells could undergo apoptosis under
non-inflammatory conditions. We then performed network reduction of the CAC network under the
non-inflammatory microenvironment and identified its attractors (Supplementary Table S4). Attractors
present the long-term behaviours of a Boolean model and can be regarded as potential stable states of
a cell under certain conditions21. In consistence with the numerical simulation results from random
initial states, two attractors, which represents the resting and apoptosis states of epithelial cells can be
identified under this condition. These simulation results can be biologically interpreted as the tendency
of IECs to remain in a resting state without inflammatory signals, but they also possess the capability
Figure 1. Topology of the CAC network. Five colours were used to represent the nodes with different
biological functions. The nodes in cyan belong to the extracellular immune microenvironment; the nodes
in orange primarily participate in inflammatory signalling; the nodes in green primarily mediate cell
proliferation; and the nodes in red regulate cell survival. The two purple nodes represent the output effects
(proliferation and apoptosis) of the network model. An arrowhead represents positive regulation (activation
or upregulation), whereas a diamond indicates negative regulation (inhibition or downregulation).
www.nature.com/scientificreports/
4
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
to undergo spontaneous apoptosis. Above simulation results are supported by previous findings that
spontaneous apoptosis of the colon epithelium is a crucial mechanism for maintaining the homeostasis
of gastrointestinal tissues22.
Dendritic cells (DCs) are known as the most potent dedicated antigen-presenting cells specialised to
initiate and maintain immunity and tolerance23. Therefore, we next simulated the normal initiation of an
inflammatory response by setting the initial state of DC to the ON state to simulate the transient activa-
tion of dendritic cells. Compared with the simulation results in the non-inflammatory microenvironment
(Fig. 2a), the transient activation of DC moderately increased the activation frequency of Proliferation
(Fig. 2b), whereas the activation frequency of Apoptosis was unchanged. In accordance, the CAC net-
work under this condition possessed attractors that represent proliferation phenotypes in addition to
apoptosis and resting phenotypes (Supplementary Table S4).The transient activation of DC also activated
STAT3, NFKB and BCATENIN, as well as other immune cells with different activation frequencies; how-
ever, none of them exceeded 0.5 (Fig. 2c and Supplementary Fig. S3). The activation frequencies of the
immune suppressive nodes, such as IL10, was generally higher than that of the pro-inflammatory nodes,
such as TNFA and IL6 (Fig. 2c). Therefore, our model suggests that the transient activation of DCs may
initiate a controlled inflammatory reaction and eventually lead to an immune suppressive microenviron-
ment, which does not support the uncontrolled growth of epithelial cells. These results are in agreement
with observations that the normal microenvironment of colon mucosa is in an immune suppressive state,
even if the colon contains a large amount of microbiota and antigens24.
Effect of different immune microenvironments on IECs.
Because the inflammatory microen-
vironment is a mixture of different types of infiltrated immune cells, we evaluated the effect of differ-
ent immune microenvironments on IECs by iteratively fixing one or a group of immune cell nodes at
the ON state to mimic their constant presence in the microenvironment. The activation frequencies of
Proliferation, Apoptosis, and different cytokine nodes in the corresponding immune microenvironment
are shown in Table 1. Because there were 63 different combinations, only the combined activations of
immune cell nodes that have additive effects to single activations are shown. We observed that in con-
trast to the transient activation of DC, maintaining DC in the ON state permanently, which could be
biologically interpreted as the constant activation of dendritic cells, created the most pro-proliferation
microenvironment, in which the activation of Proliferation significantly increased and the activation of
Apoptosis was blocked (line 1). This observation is supported by experimental findings that although the
Figure 2. Dynamics of the CAC network model in the non-inflammatory microenvironment.
(a,b) Activation frequencies for five nodes, including Proliferation, Apoptosis, STAT3, NFKB and
BCATENIN, were observed in the non-inflammatory microenvironment (a) and during transient activation
of the DC node (b,c) The stabilised activation frequencies for all the microenvironment nodes when the
initial state of DC was set to ON and other microenvironment nodes were initially set to OFF.
www.nature.com/scientificreports/
5
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
transient activation of DCs initiates a controlled inflammatory reaction24, the sustained activation of DCs
leads to chronic inflammation in IBDs25, which enhances the growth and survival of IECs7,26. In addition
to DC, the constant activation of MAC, CTL or TH1 in the microenvironment could also increase the
activation of Proliferation while decreasing the activation of Apoptosis (lines 2, 4 and 5).
However, constant activation of TREG or TH2 slightly induced the activation of Proliferation but
significantly increased the activation of Apoptosis (lines 3 and 6). Upon TREG activation, we observed
increased activation of IL10 and TGFB and decreased activation of TNFA and IL6 (line 3 compared with
lines 1 and 2). This observation is consistent with previous findings in which regulatory T cells were
shown to reduce tumour growth in CAC cases by producing immune suppressive cytokines (e.g., IL-10
and TGF-β ) and reducing pro-inflammatory cytokines (e.g., TNF-α and IL-6)27. Interestingly, although
the activation of CTL alone induced the activation of Proliferation but not Apoptosis (line 4), the com-
bined activation of CTL and TREG significantly reduced Proliferation and increased Apoptosis, forming
the most anti-tumourigenic microenvironment (line 11). This result reiterates the clinical phenomenon
that CTL contributes to intestinal inflammation and promotes tumour growth in CAC cases28, despite
of previous observations that infiltration of CTL is commonly correlated with favourable prognosis in
sporadic colon cancers29. The effect of combined activation of CTL and TREG on the states of Apoptosis
indicates that the additional activation of Treg cells can restore the cytotoxic function of CTL and there-
fore enhance immune surveillance.
We also identified several novel immune cell combinations that exhibited various effects on the states
of Proliferation, Apoptosis and the cytokine nodes (lines 7–10 and lines 12–14), revealing the complex
influence of the immune microenvironment on the survival and proliferation of IECs. These predictions
can be useful for rationally designing immune therapies to restore normal microenvironments or for
building anti-tumour microenvironments by modulating immune cells.
Dynamics of the CAC network model in a pro-tumour inflammatory microenvironment.
Notably, Proliferation did not reach full activation, even under the strongest tumour-promoting microen-
vironment (fixing DC at ON). This result could be biologically interpreted as a controlled growth of the
premalignant IECs in an inflammatory microenvironment. We then characterised the dynamic prop-
erties of the CAC network model under this tumour-promoting microenvironment to understand the
regulatory mechanisms of cellular proliferation and to identify the factors responsible for the malignant
transformation of the IECs. After a transient activation, Apoptosis was eventually stabilised in the OFF
state in all simulations, whereas the activation frequency of Proliferation was approximately 0.6 (Fig. 3a).
The activation frequencies of STAT3 and NFKB were significantly increased compared with those in the
non-inflammatory condition and the normal inflammatory response (Fig. 3a compared with Fig. 2a,b),
which is consistent with experimental observations that these two transcription factors were highly active
DC
MAC
TREG
CTL
TH1
TH2
Proliferation
Apoptosis
TNFA
IL6
TGFB
IFNG
IL10
IL12
IL4
1
•
0.66
0
0.06
1
0
0.06
0.94
1
1
2
•
0.5
0
1
1
0
0.58
0.42
1
1
3
•
0.21
0.55
0
0.21
1
0
1
0
0
4
•
0.5
0
1
1
0
1
0
1
1
5
•
0.5
0
1
1
0
1
0
1
1
6
•
0.22
0.54
0
0.22
0.78
0
1
0
1
7
•
•
0.59
0
0
1
1
0
1
1
1
8
•
•
0.5
0
1
1
0
1
0
1
1
9
•
•
•
0.43
0.57
0
1
1
1
1
1
1
10
•
•
•
•
0.5
0
1
1
1
1
1
1
1
11
•
•
0.17
0.83
0
0.17
1
1
1
0
0
12
•
•
•
0.58
0.42
0
1
0
1
1
1
1
13
•
•
•
•
0.49
0
1
1
0
1
1
1
1
14
•
•
0.17
0.83
0
0.17
0.83
1
1
0
1
Table 1. Activation frequencies of Proliferation, Apoptosis and inflammatory cytokines for different
combinatorial activations of immune cells. • indicates that the immune cell node in the header of the
corresponding column was fixed in the ON state.
www.nature.com/scientificreports/
6
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
under inflammatory stimulus7,30. BCATENIN was partially activated, although we maintained APC in
the ON state. Fixing APC in the OFF state to simulate the inactivation mutation of APC protein led to
the full activation of BCATENIN. However, the activation frequency of Proliferation was not further
increased (Supplementary Fig. S4). Clinical observations have shown that although APC mutations are
one of the earliest events in sporadic colorectal cancers and are considered essential for the transition
of preneoplastic cells to aberrant crypt foci and adenoma17,31, these mutations occur much later in CAC
cases32,33.
To further study the dynamic properties of the CAC network in the pro-tumor inflammatory microen-
vironment, we performed network reduction and analysed the attractor structure of the CAC network
under this pro-tumour microenvironment (fixing DC in the ON state). The first step of network reduc-
tion identified 33 nodes that were stabilised either in the ON or OFF state. In particular, the nodes cor-
relating with cell proliferation, such as RAS, RAF, MEK, ERK, and FOS, were stabilised in the ON state.
Previous studies have shown that ERK/MAPK (extracellular signal-regulated kinase/mitogen-activated
protein kinase) signaling can be activated by pro-inflammatory cytokines in IBD cases34 and that they
are over-activated in both sporadic colon cancer and CAC35,36. In addition, Apoptosis and other nodes
correlating with apoptotic cell death, such as BAX, TBID, CERAMIDE, CYTC, CASP3, CASP8, CASP9,
MOMP and PP2A, were all stabilised in the OFF state, indicating the entire apoptosis pathway was blocked
by the pro-tumor inflammatory microenvironment. After removing stabilised nodes, simple intermediate
nodes, and nodes with zero out-degrees, the final reduced network comprised 21 nodes and 39 interac-
tions (Fig. 3b). The Boolean rules governing the reduced network are provided in Supplementary Table
S5. We identified three attractors of the reduced CAC network model: a complex attractor (Attractor
1) that contained 48 states, and two cyclic attractors (Attractors 2 and 3) that contained six states each
(Table 2). By analysing the system’s states within these attractors, we found three activation patterns
for each node in the sub-network. One pattern was full activation, in which the node was stabilised
at ON in all states within an attractor; another pattern was partial activation, in which the node states
oscillated between ON and OFF and half were ON; and the last pattern was inactivation, in which the
node was stabilised at OFF in all states. Nodes STAT3, JAK and SOCS formed a negative feedback loop,
and they oscillated in all the attractors; however, their activation patterns were the same. Therefore,
these three nodes were excluded from further activation pattern analysis. Figure 3c shows the different
Figure 3. Dynamics of the CAC network model in the pro-tumour inflammatory microenvironment.
(a) The activation frequencies of Proliferation, Apoptosis, STAT3, NFKB and BCATENIN were observed
in the pro-tumour inflammatory microenvironment. (b) The final reduced CAC network topology in the
pro-tumour microenvironment. A green line with an arrowhead represents positive regulation, whereas a
red line with a diamond indicates negative regulation. (c) The node activation patterns in the 21-node sub-
network. Only the nodes that possess different activation patterns in the three attractors are shown. White,
grey and black boxes represent inactivation, partial activation and full activation, respectively. (d) The core
regulatory network that governed the behaviour of the CAC network in the pro-tumour inflammatory
microenvironment.
www.nature.com/scientificreports/
7
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
activation patterns of the nodes within the three attractors. The pro-proliferate nodes AKT, BCATENIN,
CYCLIND1, IKK, JUN and NFKB were all fully activated in Attractor 1, indicating a tendency for the IECs
to undergo proliferation. However, P53 and P21 were partially activated in Attractor 1. Thus, the activation
frequency of Proliferation was then restricted by the partial activation of P21 according to the Boolean
function (Proliferation* = CYCLIND1 and not P21) of the reduced CAC network. Therefore, Attractor
1 represented a limited proliferation state. In Attractor 2, AKT, BCATENIN and CYCLIND1 were fully
activated, and P21 was inactivated; thus, this attractor represented a proliferation state. Both CYCLIND1
and P21 were inactivated in Attractor 3; therefore, this attractor represented a resting state. When using
synchronous updating methods, we found the reduced network had 28 attractors, which could also be
grouped into three phenotypic classes: resting, limited proliferation and proliferation (Supplementary
Table S6).
By iteratively fixing one node in the sub-network to either an ON or OFF state, we identified a core
regulatory network that governed the behaviour of the CAC network in the pro-tumour environment
(Fig. 3d). Fixing AKT or CYCLIND1 in the OFF state could cause all state trajectories to fall into the
resting attractor. Fixing GSK3B, P21, P53 or PTEN in the ON state had the same effect on the attractor
landscape. Currently, alteration of the PTEN/PI3K/AKT signalling axis is a well-accepted driving force
in carcinogenesis, and AKT has been shown to be dysregulated in most colon cancers37,38. Notably, our
model herein emphasised that AKT activation was essential for accessing the attractor representing pro-
liferation under an inflammatory stimulus. In addition, our model suggested that GSK3-β might play a
tumour suppressor role in CAC by inhibiting CYCLIND1 and MDM2 in the pro-tumor inflammatory
microenvironment (Fig. 3d). Most importantly, we observed that only the permanent inactivation of
P53 or the activation of an endogenous P53 inhibitor, MDM2, could lead all the simulation trajectories
to fall into the proliferation attractor. This effect can be biologically interpreted as a mutation or the
constant suppression of P53, endowing IECs with the capability of uncontrolled growth and thereby
initiating malignant transformation. Therefore, the simulation results suggest P53 pathway may act as the
last guard before malignant transition in a strong pro-tumour inflammatory microenvironment, which
is supported by the observation that P53 mutation (but not APC mutation) is a common event in the
initiation phase of CAC progress39. The changes of the output effects under various conditions showed
similar trends when different updating methods were used (Supplementary Table S7), indicating the
overall dynamic properties of our Boolean network are not sensitive to the updating methods.
Key nodes regulating malignant transformation revealed by systematic node perturbations.
We subsequently performed a systematic node perturbation analysis on the entire CAC network to iden-
tify other nodes that may mediate malignant transformation in an inflammatory microenvironment.
We maintained DC in the ON state to mimic the premalignant IECs in a tumour-promoting microen-
vironment and perturbed the states of other nodes in the CAC network. For the nodes that became
stabilised in either an ON or OFF state, we fixed each node in the opposite of its stabilised state and
continued updating other nodes. For the oscillated nodes, we perturbed each node twice by fixing the
node to either ON or OFF. These perturbations mimic the manipulation of biological systems through
genetic or chemical approaches, such as gene knockdown or treating cells with active compounds. We
then observed the stabilised activation frequencies of the output nodes, Proliferation and Apoptosis, to
evaluate the perturbation effect. Through this method, we identified 36 of 109 perturbations that could
affect the activation of Proliferation or Apoptosis. We manually categorised these perturbations into
pro-proliferative, anti-proliferative, and pro-apoptosis groups according to their effects on the states of
Proliferation and Apoptosis.
As shown in Fig. 4, the pro-proliferative group contains the perturbations that lead to a high activa-
tion frequency (> 90%) of Proliferation. The perturbed nodes in this group come from four pathways:
the P53 pathway (P53, MDM2), the PI3K/AKT pathway (PI3K, AKT, PTEN, and GSK3B), the NF-κ B
pathway (NFKB, IKK, IKB) and the COX2/PGE2 pathway (PGE2, EP2 and COX2). The P53 node and
PI3K/AKT pathway nodes also exists in the anti-proliferative group when set in the opposite states
(Fig. 4). The critical roles of P53, MDM2 and AKT in CAC progress have been revealed by the above
attractor analysis. The aberrant activation of NF-κ B and COX2/PGE2 signalling has also been detected
in most CAC cases7,9. The pro-apoptotic perturbations include the inhibition of ERK MAPKs pathway
ID
Type
Length
Basin
size
Exclusive basin
size
Phenotype
1
Complex attractor
48
75%
12.5%
Limited proliferation
2
Limited cycle
6
75%
2.05%
Proliferation
3
Limited cycle
6
70%
0.03%
Resting
Table 2. Attractors of the reduced CAC network shown in Fig. 3b. The attractors can have shared basins
since the general asynchronous updating method was used. Therefore, both the total basin size and the
exclusive basin size were calculated for each attractor.
www.nature.com/scientificreports/
8
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
nodes (RAS, RAF, MEK and ERK) and the inhibition of IL6 signalling nodes (IL6 and GP130). Previous
findings indicate that ERK MAPKs are the major regulators of proliferation during colon carcinogene-
sis35, and the suppression of the ERK MAPK pathway inhibits proliferation and induce apoptosis of IECs
in an inflammatory microenvironment40. Blocking IL-6 signalling with an anti-interleukin-6 receptor
antibody or inhibiting IL-6 trans-signalling with TGF-β has also been shown to suppress tumour pro-
gression in colon cancer41. Notably, the perturbations of the nodes that participate in sphingolipid metab-
olism (CERAMIDE, SPHK1) are part of the pro-apoptotic group. A recent finding has demonstrated that
SPHK1/S1P signalling plays a crucial role in linking chronic inflammation and CAC and that inhibiting
SPHK1 could effectively reduce CAC development42.
In all, 29 of 36 predictions made by in silico perturbations can be supported by previous experi-
mental observations, which validates the rationality of our model (Supplementary Table S8). The per-
turbation analysis also led to the novel prediction that activation of GSK3B, SMAD, ROS, PP2A, ATM,
or CERAMIDE could inhibit proliferation or induce apoptosis of preneoplastic IECs in the pro-tumor
inflammatory microenvironment, indicating that these molecules can be potential therapeutic targets for
preventing CAC development.
In silico double perturbation study and experimental validations identify novel drug com-
binations.
As both our model and previous experimental studies indicate that P53 is crucial for the
malignant transformation of IECs in an inflammatory microenvironment, we maintained DC in the
ON state and P53 in the OFF state simultaneously to mimic the state of a neoplastically transformed
epithelial cell in a pro-proliferative microenvironment. We subsequently performed perturbation anal-
ysis on the CAC network model under this condition to identify potential therapeutic targets for treat-
ment of CAC. In this situation, only 18 of 89 perturbations could lower the activation frequency of
Proliferation by over 50% (Supplementary Fig. S5), indicating that the CAC network in the neoplasti-
cally transformed state was more robust than that in the pre-transformed state. Only two perturbations
could induce the activation of Apoptosis: activation of MOMP and activation of CASP3 (Supplementary
Fig. S5). However, MOMP or Caspase3 activation represent the terminal events in apoptotic cell death,
and therefore, manipulating these processes therapeutically is impractical. The robustness of the CAC
network model in the P53-inactive state suggested that the single target therapy may be less effective
in killing tumour cells that had previously developed in CAC cases. We further performed double per-
turbations by altering the state of two nodes simultaneously to seek possible combinatorial therapeutic
approaches. We found several combined perturbations that could inhibit proliferation while increasing
apoptosis (Table 3). However, not all of these perturbations are therapeutically accessible because of
the lack of modulators, such as small molecule inhibitors. Among the double perturbations, we found
some promising combinations that involve the activation of the CERAMIDE pathway while inhibiting
the PI3K/AKT pathway. The above dynamic analysis indicated that AKT may play an important role
in forming the attractor for the proliferation state, and several inhibitors of the PI3K/AKT pathway are
under clinical evaluation43. Ceramide has also been previously shown to induce apoptosis and to sensitise
tumour cells to radiotherapy44,45. Therefore, these combinations may be more clinically applicable than
other predicted combinations.
We then proceeded to validate the utility of these combinatorial perturbations in HT29 colon cancer
cells, which have a P53 R273H inactivation mutation46. The impact of the inflammatory microenviron-
ment was integrated by constantly exposing cells to the treatment with IL6 plus TNF-α , which resulted
in the activation of STAT3 and NF-κ B signalling (Supplementary Fig. S6a). As predicted, short chain,
cell permeable C2-ceramide, which has been shown to increase endogenous long chain ceramides47,
Figure 4. Node perturbation results of the CAC network model in the pre-transformed state. The
activation frequencies of node Proliferation and Apoptosis corresponding to each perturbation are shown.
(+ ) indicates that the node was fixed in the ON state, and (− ) indicates that the node was fixed in the OFF
state.
www.nature.com/scientificreports/
9
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
exerted synergistic effect with the AKT pan-inhibitor MK2206 (Fig. 5a) or PI3Kα /δ inhibitor GDC0941
(Fig. 5b) on cell viability under several concentrations, indicated by a combination index (CI) less than 1.
To further understand the mechanism of the combinational effect of AKT inhibitors and ceramide,
we extracted the sub-network related to ceramide, the PI3K/AKT pathway and inflammatory activation
from the global CAC network. According to the sub-network shown in Fig. 5c, PI3K/AKT and ceramide
signalling converged on the mitochondrial apoptotic pathway. Ceramide treatment could induce mito-
chondrial apoptosis by directly activating MOMP (mitochondria outer membrane permeabilisation) and
PP2A. MOMP could then disrupt the outer mitochondrial membrane (OMM) and mediate the subse-
quent release of death-promoting proteins, such as cytochrome C, whereas PP2A could dephosphorylate
and inactivate the anti-apoptotic protein BCL-2 and partially mediate Akt dephosphorylation. However,
when the CERAMIDE node was activated alone, the mitochondrial apoptotic pathway was inhibited
by activated AKT. AKT activated anti-apoptotic BCL-2 family proteins, such as BCL-2 and BCL-xL,
by phosphorylating and inhibiting pro-apoptotic BCL-2 family proteins, such as BAD, BIM and BAX.
Through the activation of mTOR, AKT can also inhibit PP2A and prevent the dephosphorylation and
inactivation of BCL-248. Accordingly, a combination of BCL2 inhibitor ABT263 and C2-ceramide also
had synergistic effect on cell viability (Fig. 5d). However, individual inhibition of the AKT node failed
to activate mitochondrial apoptosis due to the absence of pro-apoptotic stress, such as the activation
of CERAMIDE node. We found MOMP could only be fully activated when the CERAMIDE node was
fixed in the ON state while the AKT node remained OFF, leading to the activation of downstream
CYTC, CASP9 and CASP3, and consequently, cell apoptosis. To further validate this mechanism, we
detected apoptosis using AnnexinV-propidium iodide dual staining. In accordance with our model,
combined treatment of MK2206 and C2-ceramide showed strong synergistic apoptotic effects on HT29
cells (Fig. 5e). C2-ceramide or MK2206 alone induced approximately 20% cell apoptosis, whereas their
combination significantly increased the apoptosis level to 40–60% (Fig. 5e). These results agree with
those obtained by the combination of siRNA against AKT1, 2 and 3 and C2-ceramide (Fig. 5f and
Supplementary Fig. S6b). We consistently observed the cleavage of caspase 3, 8 and 9 and PARP follow-
ing combined treatment with MK2206 and C2-ceramide (Fig. 5g). The occurrence of apoptosis stemmed
from a key mitochondrial event, namely cytochrome C release, which was not induced by individual
treatments compared with the control group but was significantly increased after the combination of
MK2206 and C2-ceramide (Fig. 5h).
To further explore the clinical potential of our combination strategy, we tested combinations of mar-
keted chemotherapeutic drugs. Epidermal growth factor receptor (EGFR) is one of the upstream tyrosine
kinases of the PI3K/AKT pathway. Dosing HT-29 cells with C2-ceramide together with lapatinib or
gefitinib, two clinically used EGFR inhibitors, exerted synergistic effects on cell viability (Supplementary
Fig. S6c and S6d). In addition, PI3K/AKT inhibitors and FTY720 (fingolimod), which has previously
been shown to inhibit and degrade SPHK149, and conversely increase ceramide levels50, also had com-
binatory effects, although to a lesser extent than C2-ceramide (Supplementary Fig. S6e and S6f). In
agreement with our previous findings that FTY720 stimulates endogenous ceramide accumulation
by modulating sphingolipid metabolism50, FTY720 treatment elevated ceramide levels in HT-29 cells
(Supplementary Fig. S6g).
Taken together, experimental validations of the effective drug combinations and related mechanisms
in colon cancer cells further supported the rationality of our model. Most importantly, the combination
Nodes
BAX(+)
TBID(+)
FAS(+)
CYTC(+)
CASP8(+)
CASP9(+)
CERAMIDE(+)
AKT(− )
0.06/0.4
0.06/0.41
0.06/0.68
0.05/0.34
0.06/0.69
0.06/0.47
0/1
PI3K(− )
0.06/0.4
0.06/0.41
0.07/0.67
0.06/0.34
0.07/0.68
0.06/0.47
0/1
PTEN(+ )
0.06/0.4
0.06/0.41
0.06/0.68
0.05/0.35
0.07/0.68
0.06/0.47
0/1
PP2A(+ )
0/1
0/1
0.06/0.94
0.06/0.34
0/1
006/0.47
0/1
BCL2(− )
0/1
0/1
NA
NA
0/1
NA
0/1
RAS(− )
0/0.41
0/0.41
0/0.84
0/0.35
0/0.69
0/0.48
0.06/0.94
IL6(− )
0/1
0/1
0/1
0/1
0/1
0/1
0/1
GP130(− )
0.06/0.94
0.06/0.91
0.06/0.94
0.05/0.95
0.05/0.95
0.06/0.94
0.06/0.94
IKK(− )
0.37/0.45
0.33/0.46
0.12/0.72
0.4/0.38
0.12/0.72
0.25/0.51
0/1
IKB(+ )
0.33/0.46
0.33/0.46
0.1/0.73
0.38/0.38
0.1/0.73
0.22/0.52
0/1
NFKB(− )
0.33/0.46
0.33/0.46
0.1/0.73
0.37/0.39
0.1/0.73
0.22/0.51
0/1
IAP(− )
NA
NA
NA
0/1
0/1
0/1
NA
Table 3. Double perturbations that may sensitise IECs to pro-apoptotic signals. (+ ) indicates fixing the
node in the ON state (activation), (− ) indicates fixing the node in the OFF state (inhibition). The results are
shown as the activation frequency of Proliferation/the activation frequency of Apoptosis, and NA indicates
no additional effect on Apoptosis and Proliferation compared with single perturbation.
www.nature.com/scientificreports/
10
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
strategy provides a mechanism-based rational therapeutic approach for CAC, as well as for other
inflammation-associated cancers.
Discussion
In this article, we presented a reconstruction of the CAC network and its implementation as a discrete
dynamic Boolean model. The rationality of the CAC network model was justified by comparing the
simulation results with existing observations, as well as novel experimental validations. Comprehensive
analysis of the dynamic properties of the CAC network was also performed to unravel the missing link
between chronic inflammation and cancer development and to identify potential therapeutic targets.
Figure 5. Experimental validations of drug combination predictions that targeting AKT plus exogenous
C2-ceramide reduced cell viability and increased cell apoptosis. (a) The combination effects of C2-
ceramide and MK2206 on the viability of HT29 cells were determined by calculating the CI values for
each data point. CI < 1 indicates a synergistic effect. (b) The combination effects of C2-ceramide and
GDC0941 on the viability of HT29 cells. (c) The sub-network related to PI3K/AKT and ceramide signalling
extracted from the entire CAC network. A green line with an arrowhead indicates positive regulation
(activation or upregulation), whereas a red line with a diamond indicates negative regulation (inhibition or
downregulation). (d) The combination effects of C2-ceramide and ABT263 on the viability of HT29 cells.
(e) Synergistic apoptotic effects of C2-ceramide and MK2206 on HT29 cells. (f) Mean percentage of
apoptotic cells treated with siAKT1/2/3 and/or 15 μ M C2-ceramide. ‘NC’ group stands for the scrambled
negative siRNA pools, which was used as a negative control; ‘#1’ and ‘#2’ groups stand for AKT siRNA pools
that contain different siRNA sequences listed in Supplementary Methods. (g) Immunoblots of lysates from
cells treated with 15 μ M C2-ceramide and 10 μ M or 15 μ M MK2206 for 24 h. (h) Flow cytometry detection
of mitochondrial cytochrome C levels in HT29 cells treated with DMSO (red line), 15 μ M C2-ceramide
(blue line), 10 μ M MK2206 (orange line) and 15 μ M C2-ceramide+ 10 μ M MK2206 (green line) for 24 h.
The bar graphs (right) show the relative fluorescence intensities representing mitochondrial cytochrome C
levels. Data are representative of three independent experiments (mean ± s.e.m.). **P < 0.01 and ***P < 0.001
(Student’s t-test). ns, not significant.
www.nature.com/scientificreports/
11
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
Through manipulation of the nodes in the microenvironment component of the CAC network model,
we suggests that dendritic cells play critical roles in forming the pro-proliferative inflammatory microen-
vironment. The transient activation of DCs initiates a controlled inflammatory reaction and eventually
leads to an immune suppressive microenvironment, whereas the sustained activation of DCs leads to a
dysregulated inflammatory microenvironment, which strongly supports the proliferation and survival
of IECs. In addition, we thoroughly characterized the dynamic properties of CAC network under the
pro-tumor inflammatory microenvironment and identified a core regulatory network that governed the
cell outcome. According to the core regulatory network, P53 inactivation was found to be critical for
malignant transformation of epithelial cells under the pro-tumor inflammatory microenvironment. The
result was supported by current findings of a dysregulated P53 pathway during CAC development and
partly explained that in the ‘two-hit’ model, a somatic mutation is necessary for tumour initiation in a
pro-tumour inflammatory microenvironment39.
Subsequent systematic perturbation analysis indicated that various dysregulations could facilitate
the malignant transformation of preneoplastic IECs in an inflammatory microenvironment, including
the hyperactivation of the NF-κ B, PI3K/AKT or the COX2 pathways. Many perturbations were also
predicted to suppress cell survival and proliferation of IECs in an inflammatory microenvironment,
such as the inhibition of the ERK/MAPK pathway or the SPHK1/S1P pathway. These perturbations
can be used as an early intervention method to prevent CAC development. However, the CAC network
in a P53 inactive state, which mimicked malignant transformed IECs, was more resistant to perturba-
tions. Double perturbation studies on the P53-inactivated CAC network suggested that combinatorial
intervention methods through multi-targeted drugs or drug combinations could be more effective at
treating later-stage CAC patients. Most importantly, we discovered and validated some novel combina-
torial therapeutic approaches. We found that simultaneously inhibiting PI3K/AKT signalling and add-
ing C2-ceramide, a pro-apoptotic sphingolipid signalling molecule, had a synergistic cytotoxic effect
on colon cancer cells under an inflammatory stimulus. We explored the underlying mechanism of this
synergistic effect by combining biochemical experiments and network simulations. We demonstrated
that this effect was primarily attributed to regulation of the mitochondrial apoptotic pathway. As a
pro-apoptotic signalling molecule, ceramide can directly or indirectly target mitochondria and lead
to the release of apoptotic proteins, such as cytochrome C, and initiate the apoptosis machinery51,52.
However, in many tumour cells, the PI3K/AKT pathway is highly active, which could in turn adversely
affect the integrity of the mitochondria outer membrane by indirectly activating anti-apoptotic BCL-2
family proteins, such as BCL-2 or BCL-xL53. Our simulation results and previous experimental findings
also suggest that inflammatory signalling can activate the AKT pathway to enhance survival of tumour
cells. Therefore, only when PI3K/AKT signalling is blocked can the apoptosis machinery effectively be
activated by a pro-apoptotic stimulus, such as ceramide (Fig. 6). Our results may also explain previous
findings in which D, L-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP), a modula-
tor of ceramide metabolism that elevates endogenous ceramide levels, could sensitise leukemic cells to
the treatment of ABT263, an inhibitor of anti-apoptotic BCL2-like proteins54.
Figure 6. Proposed model for the synergistic effect between ceramide and PI3K/AKT pathway inhibitors
in inducing apoptotic cell death of cancer cells. Ceramide directly or indirectly targets mitochondria,
leading to the release of apoptotic proteins, such as cytochrome C, and activates caspases. However, if the
PI3K/AKT pathway is activated by an inflammatory stimulus or other growth factors, it can preserve the
integrity of the mitochondrial outer membrane by activating the anti-apoptotic BCL-2 family of proteins,
such as BCL-2 or BCL-xL, by inhibiting PP2A or BAD. Therefore, ceramide and PI3K/AKT pathway
inhibitors exert a synergistic cytotoxic effect on cancer cells. Note: the arrows in this simplified scheme are
not intended to indicate direct physiological interactions.
www.nature.com/scientificreports/
12
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
We are aware that this model is unable to capture the full complexity of CAC development. Some
unknown factors may be present in CAC development, and merely incorporating outcomes, such as
proliferation and apoptosis, is insufficient to assess tumour development. However, the basic property of
tumour cells is uncontrolled proliferation, and inducing growth arrest or apoptosis is the most effective
method to prevent cancer development. Therefore, our theoretical model can provide valuable informa-
tion to guide further experimental studies, and this model can be easily refined and expanded with the
availability of additional information.
In conclusion, the dynamic modelling of the CAC development process can lead to a better mechanis-
tic understanding of CAC and other inflammation-associated cancers because CAC serves as a paradigm
for inflammation-associated cancer development. Our model and experimental findings will also be
helpful in identifying novel therapeutic targets and the design of combinatorial therapeutic approaches
to achieve early prevention and treatment of CAC.
Materials and Methods
Construction of the CAC network.
A hierarchical method was used to construct the CAC network.
First, signalling components that are critical participants in IBD and colorectal cancer, as indicated by a
literature search, were collected. These components were used as seed nodes to build the initial network
model. Then, the initial network was expanded using the GeneGo database (http://www.genego.com/).
The regulatory relationships were then verified manually, and the interactions that were not specifically
mentioned to be relevant to colitis or colon cancer were removed. Further, a similar approach, such as the
one Zhang et al.13,55 used in constructing T-LGL network model, was adopted to remove the redundant
and indirect interactions between nodes.
Boolean dynamic modelling.
In the Boolean model, each node has only two discrete states: ON and
OFF (1 and 0). The regulatory relationships between upstream nodes (regulators) and downstream nodes
(targets) are expressed by the logical operators AND, OR and NOT56. The future state of each node can
be determined by the current states of the nodes and the Boolean transfer function
,
→
,
{
}
f : {0 1}
0 1
i
ki
,
where ki is the number of regulators of node i. In Boolean models, the time variable is discrete and
usually designated as steps. To propagate the discrete states of a Boolean model, different node-updating
strategies have been proposed, such as synchronous and asynchronous update methods21. In this study,
we mainly used a general asynchronous (GA) method57, in which a randomly selected node is updated
at each time step. To evaluate the general behaviour trends for all nodes in the network model, multiple
replicate simulations were performed from the same initial conditions but with random update orders.
These trends can be reflected by the activation frequency for each node, which was calculated by dividing
the number of simulations where the node is ON by the total replicate number. The GA method has been
widely used in modelling signal transduction networks and has been suggested to be the most informa-
tive and computationally efficient method among asynchronous updating strategies21,56. We also com-
pared the simulation results using synchronous method, GA method and another asynchronous updating
method—random order asynchronous (ROA) method58.
Network reduction.
Considering that the size of the state space of a Boolean model is exponentially
dependent on the node number (a Boolean network with n nodes has 2n states) and attractor identifica-
tion is a strong NP-complete problem, tracking all of the attractors within a relatively small network is
computationally demanding. Therefore, we used a network reduction method proposed by Saadatpour
et al.21 to reduce the node number while maintaining the long-term behaviour of the dynamic model.
First, the nodes that stabilise in an attracting state (ON or OFF) during the entire simulation are elimi-
nated. The attracting states of these nodes are only determined by the states of the input nodes and can
be readily identified by inspecting the Boolean functions. Second, the simple mediator nodes, with both
in-degrees and out-degrees equal to one, are iteratively removed, and their input and output nodes are
connected directly. The dangling nodes (nodes with zero out-degrees) are also removed. This method
can effectively reduce the network size and maintain the fixed point, as well as the complex attractor of
Boolean models, using either synchronous or GA methods.
Attractor identification.
When updating a Boolean model, the state of the whole system at a certain
time step is defined by the collection of the states of all the nodes at that step. As the Boolean model
evolves over time, all the possible states of the system constitute the state space, which can be represented
by a state transition graph whose nodes are the states of the system and the edges are allowed transitions
among states. Attractors are special states in the state transition graph that the system will eventually
settle down to and will not transit to other states. An attractor can either be a fixed point, in which the
state of the system does not change, or a complex attractor, in which the states traverse regularly or
irregularly over a series of states56.
To identify the attractors of the reduced CAC network model, we firstly constructed the state transi-
tion graph by updating the model starting from all possible initial states. Subsequently, the strongly con-
nected components (SCCs) of the state transition graph were identified. SCCs are subgraphs of a directed
graph in which every node is reachable from every other node. Complex attractors of a Boolean model
www.nature.com/scientificreports/
13
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
are the SCCs with empty out-component of the state transition graph59. Fixed point attractors can also
be identified by this approach because a fixed point can be regarded as an SCC that contains only one
state and is strongly connected to itself. The states in the state transition graph that can reach a certain
attractor were marked as the basin states of that attractor.
Coding.
We implemented the functions used in this study into an open source Python software
package named SimpleBool, which can be downloaded at https://github.com/SimpleBool/SimpleBool.
SimpleBool directly reads model and parameter input files to perform dynamic simulations, attractor
identification and in silico perturbation studies. SimpleBool can run in a stand-alone mode, and there-
fore, coding experience is not required. Readers can refer to the website for further guidance on how to
use the software.
Antibody and compounds.
Antibodies against STAT3, p-STAT3 (Y705), P65, p-P65 (S536), AKT,
p-AKT (S473), PARP, cleaved-caspase 3, cleaved-caspase 8 and cleaved-caspase 9 were obtained from Cell
Signalling Technology. Anti-GAPDH antibody was from Epitomics. MK2206, GDC0941 and ABT263
were purchased from Selleckchem.
Cell line, cell culture and in vitro experiments.
HT29 cells were obtained from the American
Type Culture Collection (Rockville, USA). Cell line identity is routinely monitored by short tandem
repeat (STR) analysis. Cell lines were grown at 37 °C in a 5% CO2 incubator. The cell medium was
McCoy’s 5a (Gibco) supplemented with 10% FBS; 50 U/ml penicillin, 50 U/ml streptomycin.50 ng/ml IL6
(PeproTech) and 10 ng/ml TNF-α (PeproTech) were added to the medium to model an inflammatory
microenvironment. Cell viability was measured using the Cell Proliferation Reagent sulforhodamine B
(SRB, sigma). For siRNA transfection, cells were plated at 3 × 105 cells/ml in OPTI-MEM serum-free
medium and transfected with a specific siRNA duplex using Lipofectamine® RNAiMAX Reagent Agent
(Life Technologies) according to the manufacturer’s instructions. Synergy between combination treat-
ments was determined by the combination index using CompuSyn software, available online.
Flow cytometry.
Cells were plated and treated the following day with the indicated agents. Cells were
detached using trypsin-EDTA, resuspended in growth medium and counted. To detect cell apoptosis
using Annexin V/propidium iodide staining, 1 × 106 cells were washed with cold PBS, resuspended in
100 μ l of binding buffer, and then, propidium iodide and FITC-labelled antibody against annexin V were
added according to the manufacturer’s protocol (Vazyme Biotech). FlowCellect Cytochrome c Reagents
(Millipore) were obtained for analysis of cytochrome C release from mitochondria. Briefly, 1 × 105 cells
were washed with PBS, and then, a permeabilsation buffer working solution and a fixation buffer work-
ing solution were added sequentially to achieve selective permeabilisation of mitochondria while leaving
the mitochondrial membrane intact. Finally, 10 μ l of either the anti-IgG1-FITC isotype control or the
anti-cytochrome c-FITC antibody was added to each sample according to the manufacturer’s protocol.
At least 3 × 103 cells per sample were analysed with a FACScan flow cytometer (Becton Dickinson).
Statistical analysis.
Data are representative of three independent experiments (mean ± s.e.m.). A
two-tailed Student’s t-test was used for statistical comparisons between groups, and P-values ≤ 0.05 were
considered statistically significant.
References
1. Balkwill, F. & Mantovani, A. Inflammation and cancer: back to Virchow? Lancet 357, 539–545, doi: 10.1016/S0140-6736(00)04046-0
(2001).
2. Garagnani, P., Pirazzini, C. & Franceschi, C. Colorectal cancer microenvironment: among nutrition, gut Microbiota, inflammation
and epigenetics. Curr Pharm Design 19, 765–778 (2013).
3. Ekbom, A., Helmick, C., Zack, M. & Adami, H. O. Increased risk of large-bowel cancer in Crohn’s disease with colonic
involvement. Lancet 336, 357–359 (1990).
4. Giardiello, F. M., Offerhaus, G. J. & DuBois, R. N. The role of nonsteroidal anti-inflammatory drugs in colorectal cancer
prevention. Eur J Cancer 31A, 1071–1076 (1995).
5. Lin, W. W. & Karin, M. A cytokine-mediated link between innate immunity, inflammation, and cancer. J Clin Invest 117,
1175–1183, doi: 10.1172/Jci31537 (2007).
6. Yu, H., Kortylewski, M. & Pardoll, D. Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment.
Nat Rev Immunol 7, 41–51, doi: 10.1038/Nri1995 (2007).
7. Greten, F. R. et al. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118,
285–296, doi: 10.1016/j.cell.2004.07.013 (2004).
8. Bierie, B. & Moses, H. L. Transforming growth factor beta (TGF-beta) and inflammation in cancer. Cytokine & growth factor
reviews 21, 49–59, doi: 10.1016/j.cytogfr.2009.11.008 (2010).
9. Wang, D. & Dubois, R. N. The role of COX-2 in intestinal inflammation and colorectal cancer. Oncogene 29, 781–788, doi:
10.1038/onc.2009.421 (2010).
10. de Visser, K. E. & Jonkers, J. Towards understanding the role of cancer-associated inflammation in chemoresistance. Curr Pharm
Des 15, 1844–1853 (2009).
11. Choi, M., Shi, J., Jung, S. H., Chen, X. & Cho, K. H. Attractor landscape analysis reveals feedback loops in the p53 network that
control the cellular response to dNA damage. Sci Signal 5, ra83, doi: 10.1126/scisignal.2003363 (2012).
12. Singh, A., Nascimento, J. M., Kowar, S., Busch, H. & Boerries, M. Boolean approach to signalling pathway modelling in HGF-
induced keratinocyte migration. Bioinformatics 28, I495–I501, doi: 10.1093/bioinformatics/bts410 (2012).
www.nature.com/scientificreports/
14
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
13. Zhang, R. R. et al. Network model of survival signaling in large granular lymphocyte leukemia. Proceedings of the National
Academy of Sciences of the United States of America 105, 16308–16313, doi: 10.1073/pnas.0806447105 (2008).
14. Naldi, A., Carneiro, J., Chaouiya, C. & Thieffry, D. Diversity and plasticity of Th cell types predicted from regulatory network
modelling. PLoS computational biology 6, e1000912, doi: 10.1371/journal.pcbi.1000912 (2010).
15. Saez-Rodriguez, J. et al. Comparing signaling networks between normal and transformed hepatocytes using discrete logical
models. Cancer research 71, 5400–5411 (2011).
16. Fumia, H. F. & Martins, M. L. Boolean network model for cancer pathways: predicting carcinogenesis and targeted therapy
outcomes. Plos One 8, e69008, doi: 10.1371/journal.pone.0069008 (2013).
17. Morin, P. J. et al. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 275,
1787–1790 (1997).
18. Kojima, M. et al. Increased nuclear factor-kB activation in human colorectal carcinoma and its correlation with tumor progression.
Anticancer Res 24, 675–681 (2004).
19. Atreya, R. & Neurath, M. F. Involvement of IL-6 in the pathogenesis of inflammatory bowel disease and colon cancer. Clin Rev
Allergy Immunol 28, 187–196 (2005).
20. Yu, H., Pardoll, D. & Jove, R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer 9, 798–809,
doi: 10.1038/Nrc2734 (2009).
21. Saadatpour, A., Albert, I. & Albert, R. Attractor analysis of asynchronous Boolean models of signal transduction networks.
Journal of theoretical biology 266, 641–656, doi: 10.1016/j.jtbi.2010.07.022 (2010).
22. Potten, C. S. The significance of spontaneous and induced apoptosis in the gastrointestinal tract of mice. Cancer Metastasis Rev
11, 179–195 (1992).
23. Rescigno, M. & Di Sabatino, A. Dendritic cells in intestinal homeostasis and disease. J Clin Invest 119, 2441–2450, doi: 10.1172/
JCI39134 (2009).
24. Fiocchi, C. The normal intestinal mucos: a state of ‘controlled inflammation’ in Inflammatory bowel disease: from bench to bedside
(ed Fergus Shanahan Loren C. Karp, Stephan R. Targan) Ch. 8, 101–120 (Springer, 2005).
25. Hart, A. L. et al. Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology 129, 50–65 (2005).
26. MacDonald, T. T. Cytokine regulation of intestinal epithelial cell proliferation. Expert Rev Clin Immunol 6, 531–535, doi: 10.1586/
eci.10.42 (2010).
27. Erdman, S. E. et al. CD4+ CD25+ regulatory lymphocytes induce regression of intestinal tumors in ApcMin/+ mice. Cancer Res
65, 3998–4004 (2005).
28. Waldner, M. J. & Neurath, M. F. Colitis-associated cancer: the role of T cells in tumor development. Semin Immunopathol 31,
249–256, doi: 10.1007/s00281-009-0161-8 (2009).
29. Naito, Y. et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res
58, 3491–3494 (1998).
30. Grivennikov, S. et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated
cancer. Cancer Cell 15, 103–113, doi: 10.1016/j.ccr.2009.01.001 (2009).
31. Redston, M. S., Papadopoulos, N., Caldas, C., Kinzler, K. W. & Kern, S. E. Common occurrence of APC and K-ras gene mutations
in the spectrum of colitis-associated neoplasias. Gastroenterology 108, 383–392 (1995).
32. Tarmin, L. et al. Adenomatous polyposis coli gene mutations in ulcerative colitis-associated dysplasias and cancers versus
sporadic colon neoplasms. Cancer Res 55, 2035–2038 (1995).
33. Aust, D. E. et al. The APC/beta-catenin pathway in ulcerative colitis-related colorectal carcinomas: a mutational analysis. Cancer-
Am Cancer Soc 94, 1421–1427 (2002).
34. Broom, O. J., Widjaya, B., Troelsen, J., Olsen, J. & Nielsen, O. H. Mitogen activated protein kinases: a role in inflammatory bowel
disease? Clin Exp Immunol 158, 272–280 (2009).
35. Fang, J. Y. & Richardson, B. C. The MAPK signalling pathways and colorectal cancer. Lancet Oncol 6, 322–327 (2005).
36. Bos, J. L. et al. Prevalence of ras gene mutations in human colorectal cancers. Nature 327, 293–297, doi: 10.1038/327293a0
(1987).
37. Itoh, N. et al. Phosphorylation of Akt/PKB is required for suppression of cancer cell apoptosis and tumor progression in human
colorectal carcinoma. Cancer-Am Cancer Soc 94, 3127–3134, doi: 10.1002/Cncr.10591 (2002).
38. Khan, M. W. et al. PI3K/AKT signaling is essential for communication between tissue-infiltrating mast cells, macrophages, and
epithelial cells in colitis-induced cancer. Clin Cancer Res 19, 2342–2354, doi: 10.1158/1078-0432.Ccr-12-2623 (2013).
39. Rubin, D. C., Shaker, A. & Levin, M. S. Chronic intestinal inflammation: inflammatory bowel disease and colitis-associated colon
cancer. Front Immunol 3, 107, doi: 10.3389/fimmu.2012.00107 (2012).
40. Yan, F. et al. Kinase suppressor of Ras-1 protects intestinal epithelium from cytokine-mediated apoptosis during inflammation.
J Clin Invest 114, 1272–1280, doi: 10.1172/JCI21022 (2004).
41. Becker, C. et al. TGF-beta suppresses tumor progression in colon cancer by inhibition of IL-6 trans-signaling. Immunity 21,
491–501, doi: 10.1016/j.immuni.2004.07.020 (2004).
42. Liang, J. et al. Sphingosine-1-phosphate links persistent STAT3 activation, chronic intestinal inflammation, and development of
colitis-associated cancer. Cancer Cell 23, 107–120, doi: 10.1016/j.ccr.2012.11.013 (2013).
43. Martini, M., De Santis, M. C., Braccini, L., Gulluni, F. & Hirsch, E. PI3K/AKT signaling pathway and cancer: an updated review.
Annals of medicine 46, 372–383, doi: 10.3109/07853890.2014.912836 (2014).
44. Haimovitz-Friedman, A., Kolesnick, R. N. & Fuks, Z. Ceramide signaling in apoptosis. British medical bulletin 53, 539–553
(1997).
45. Litvak, D. A., Bilchik, A. J. & Cabot, M. C. Modulators of ceramide metabolism sensitize colorectal cancer cells to chemotherapy:
a novel treatment strategy. Journal of gastrointestinal surgery: official journal of the Society for Surgery of the Alimentary Tract 7,
140–148 (2003).
46. Ahmed, D. et al. Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis 2, e71, doi: 10.1038/oncsis.2013.35
(2013).
47. Ogretmen, B. et al. Biochemical mechanisms of the generation of endogenous long chain ceramide in response to exogenous
short chain ceramide in the A549 human lung adenocarcinoma cell line. Role for endogenous ceramide in mediating the action
of exogenous ceramide. J Biol Chem 277, 12960–12969, doi: 10.1074/jbc.M110699200 (2002).
48. Hay, N. & Sonenberg, N. Upstream and downstream of mTOR. Genes & development 18, 1926–1945, doi: 10.1101/gad.1212704
(2004).
49. Lim, K. G. et al. FTY720 analogues as sphingosine kinase 1 inhibitors: enzyme inhibition kinetics, allosterism, proteasomal
degradation, and actin rearrangement in MCF-7 breast cancer cells. J Biol Chem 286, 18633–18640, doi: 10.1074/jbc.M111.220756
(2011).
50. Chen, L. et al. FTY720 induces apoptosis of M2 subtype acute myeloid leukemia cells by targeting sphingolipid metabolism and
increasing endogenous ceramide levels. Plos One 9, e103033, doi: 10.1371/journal.pone.0103033 (2014).
51. Colombini, M. Ceramide channels and their role in mitochondria-mediated apoptosis. Bba-Bioenergetics 1797, 1239–1244, doi:
10.1016/j.bbabio.2010.01.021 (2010).
www.nature.com/scientificreports/
15
Scientific Reports | 5:14739 | DOI: 10.1038/srep14739
52. Dobrowsky, R. T., Kamibayashi, C., Mumby, M. C. & Hannun, Y. A. Ceramide activates heterotrimeric protein phosphatase 2A.
J Biol Chem 268, 15523–15530 (1993).
53. Stiles, B. L. PI-3-K and AKT: Onto the mitochondria. Advanced drug delivery reviews 61, 1276–1282, doi: 10.1016/j.
addr.2009.07.017 (2009).
54. Casson, L. et al. Inhibition of ceramide metabolism sensitizes human leukemia cells to inhibition of BCL2-like proteins. Plos One
8, e54525, doi: 10.1371/journal.pone.0054525 (2013).
55. Kachalo, S., Zhang, R., Sontag, E., Albert, R. & DasGupta, B. NET-SYNTHESIS: a software for synthesis, inference and
simplification of signal transduction networks. Bioinformatics 24, 293–295, doi: 10.1093/bioinformatics/btm571 (2008).
56. Albert, I., Thakar, J., Li, S., Zhang, R. & Albert, R. Boolean network simulations for life scientists. Source code for biology and
medicine 3, 16, doi: 10.1186/1751-0473-3-16 (2008).
57. Harvey, I. & Bossomaier, T. Time out of joint: Attractors in asynchronous random boolean networks (MIT Press, Cambridge, 1997).
58. Chaves, M., Albert, R. & Sontag, E. D. Robustness and fragility of Boolean models for genetic regulatory networks. Journal of
theoretical biology 235, 431–449, doi: 10.1016/j.jtbi.2005.01.023 (2005).
59. Saadatpour, A. & Albert, R. Boolean modeling of biological regulatory networks: a methodology tutorial. Methods 62, 3–12,
doi: 10.1016/j.ymeth.2012.10.012 (2013).
Acknowledgements
We are extremely grateful to Dr. Sarah Spiegel for the great discussion and suggestion which significantly
improve the quality of our manuscript. We gratefully acknowledge financial support from the Hi-
Tech Research and Development Program of China (2012AA020302 to CL), the Ministry of Science
and Technology of China (2015CB910304 to HL), the National Natural Science Foundation of China
(91029704 to CL, 91229204 to HL, 81302700 to ZL, 21472208 to CL, 81230076 and 21210003 to HJ).
Author Contributions
C.L., M.H. and M.G. conceived the project. J.L., Z.L., H.L.Z.(Hanlin Zeng), M.H., M.G. and C.L. designed
the experiments. J.L. and Z.L. completed the coding part. L.C., L.Z. and H.Z. (Hao Zhang) collected data
from literature and database. J.L., H.J., Z.L. and C.L. designed and performed computational studies and
analysed the data. H.L.Z. and M.H. performed all cell-based assays. J.L., Z.L., H.L.Z., M.H., H.L., B.S. and
C.L. analysed the data and wrote the manuscript. S.S. provided suggestions and helped with manuscript
editing. All authors read and approved the final manuscript.
Additional Information
Supplementary information accompanies this paper at http://www.nature.com/srep
Competing financial interests: The authors declare no competing financial interests.
How to cite this article: Lu, J. et al. Network modelling reveals the mechanism underlying colitis-
associated colon cancer and identifies novel combinatorial anti-cancer targets. Sci. Rep. 5, 14739;
doi: 10.1038/srep14739 (2015).
This work is licensed under a Creative Commons Attribution 4.0 International License. The
images or other third party material in this article are included in the article’s Creative Com-
mons license, unless indicated otherwise in the credit line; if the material is not included under the
Creative Commons license, users will need to obtain permission from the license holder to reproduce
the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
|
26446703
|
ROS = ( ( TNFR ) AND NOT ( SOD ) )
MEK = ( RAF ) OR ( ROS )
Proliferation = ( ( ( CYCLIND1 AND ( ( ( FOS ) ) ) ) AND NOT ( P21 ) ) AND NOT ( CASP3 ) )
SMAD7 = ( NFKB ) OR ( SMAD )
SMAD = ( ( TGFR ) AND NOT ( JUN ) )
CFLIP = ( NFKB )
PP2A = ( ( CERAMIDE ) AND NOT ( AKT ) )
TH1 = ( ( ( ( IFNG ) AND NOT ( IL4 ) ) AND NOT ( IL10 ) ) AND NOT ( TGFB ) ) OR ( ( ( ( IL12 ) AND NOT ( IL4 ) ) AND NOT ( IL10 ) ) AND NOT ( TGFB ) )
JNK = ( MEKK1 ) OR ( ASK1 )
BCATENIN = NOT ( ( APC AND ( ( ( GSK3B ) ) ) ) )
TNFR = ( TNFA )
IL10 = ( TH2 ) OR ( TREG )
TGFB = ( TREG )
IFNG = ( CTL ) OR ( TH1 )
STAT3 = ( JAK )
RAS = ( EP2 ) OR ( GP130 )
MOMP = ( ( CERAMIDE ) AND NOT ( BCL2 ) ) OR ( ( BAX ) AND NOT ( BCL2 ) ) OR ( ( TBID ) AND NOT ( BCL2 ) )
DC = ( ( TNFA ) AND NOT ( IL10 ) ) OR ( ( CCL2 ) AND NOT ( IL10 ) )
CERAMIDE = ( ( SMASE ) AND NOT ( SPHK1 ) )
IL6 = ( MAC ) OR ( DC ) OR ( NFKB )
CASP9 = ( ( ( CYTC ) AND NOT ( P21 ) ) AND NOT ( IAP ) )
Apoptosis = ( CASP3 )
ATM = ( ROS )
CYTC = ( MOMP )
P53 = ( ( PTEN ) AND NOT ( MDM2 ) ) OR ( ( ATM ) AND NOT ( MDM2 ) ) OR ( ( JNK ) AND NOT ( MDM2 ) )
SMAC = ( MOMP )
CTL = ( ( IFNG ) AND NOT ( TGFB ) )
COX2 = ( TNFR AND ( ( ( S1P ) ) ) )
CASP3 = ( ( CASP9 ) AND NOT ( IAP ) ) OR ( ( CASP8 ) AND NOT ( IAP ) )
ERK = ( MEK )
GSK3B = NOT ( ( AKT ) OR ( EP2 ) )
FOS = ( ERK )
TREG = ( ( IL10 ) AND NOT ( IL6 ) ) OR ( ( DC ) AND NOT ( IL6 ) )
MEKK1 = ( TGFR ) OR ( CERAMIDE ) OR ( TNFR )
NFKB = NOT ( ( IKB ) )
FADD = ( FAS ) OR ( TNFR )
TNFA = ( MAC )
BCL2 = ( ( ( STAT3 ) AND NOT ( P53 ) ) AND NOT ( PP2A ) ) OR ( ( ( NFKB ) AND NOT ( P53 ) ) AND NOT ( PP2A ) )
CYCLIND1 = ( ( BCATENIN ) AND NOT ( GSK3B ) ) OR ( ( JUN ) AND NOT ( GSK3B ) ) OR ( ( STAT3 ) AND NOT ( GSK3B ) )
JAK = ( ( GP130 ) AND NOT ( SOCS ) )
PTEN = ( ( ( P53 ) AND NOT ( JUN ) ) AND NOT ( NFKB ) )
SMASE = ( FADD ) OR ( P53 )
IL4 = ( DC ) OR ( TH2 )
MDM2 = ( ( ( P53 AND ( ( ( AKT ) ) ) ) AND NOT ( ATM ) ) AND NOT ( GSK3B ) )
TBID = ( ( CASP8 ) AND NOT ( BCL2 ) )
JUN = ( ( BCATENIN AND ( ( ( JNK ) ) ) ) AND NOT ( GSK3B ) ) OR ( ( ERK AND ( ( ( JNK ) ) ) ) AND NOT ( GSK3B ) )
S1P = ( SPHK1 )
SPHK1 = ( TNFR ) OR ( ERK )
MAC = ( ( IFNG ) AND NOT ( IL10 ) ) OR ( ( CCL2 ) AND NOT ( IL10 ) )
TGFR = ( ( TGFB ) AND NOT ( SMAD7 ) )
PGE2 = ( COX2 )
RAF = ( CERAMIDE ) OR ( RAS )
SOCS = ( STAT3 )
P21 = ( ( ( P53 ) AND NOT ( CASP3 ) ) AND NOT ( GSK3B ) ) OR ( ( ( SMAD ) AND NOT ( CASP3 ) ) AND NOT ( GSK3B ) )
BAX = ( ( TBID AND ( ( ( PP2A ) ) ) ) AND NOT ( AKT ) ) OR ( ( P53 AND ( ( ( PP2A ) ) ) ) AND NOT ( AKT ) )
IKK = ( AKT ) OR ( TNFR AND ( ( ( S1P ) ) ) )
FAS = ( CTL )
CCL2 = ( NFKB )
EP2 = ( PGE2 )
SOD = ( STAT3 ) OR ( NFKB )
PI3K = ( ( EP2 ) AND NOT ( PTEN ) ) OR ( ( RAS ) AND NOT ( PTEN ) )
AKT = ( ( ( PI3K ) AND NOT ( CASP3 ) ) AND NOT ( PP2A ) )
IL12 = ( MAC ) OR ( DC )
CASP8 = ( ( ( FADD ) AND NOT ( P21 ) ) AND NOT ( CFLIP ) )
ASK1 = ( ( ROS ) AND NOT ( P21 ) )
IKB = NOT ( ( IKK ) )
GP130 = ( IL6 )
TH2 = ( ( ( IL4 ) AND NOT ( IFNG ) ) AND NOT ( TGFB ) )
IAP = ( ( NFKB ) AND NOT ( SMAC ) ) OR ( ( STAT3 ) AND NOT ( SMAC ) )
|
RESEARCH ARTICLE
Mathematical Modelling of Molecular
Pathways Enabling Tumour Cell Invasion and
Migration
David P. A. Cohen1,2,3, Loredana Martignetti1,2,3, Sylvie Robine1,4, Emmanuel Barillot1,2,3,
Andrei Zinovyev1,2,3‡, Laurence Calzone1,2,3‡*
1 Institut Curie, Paris, France, 2 INSERM, U900, Paris, France, 3 Mines ParisTech, Fontainebleau, Paris,
France, 4 CNRS UMR144, Paris, France
‡ These authors are joint senior authors on this work.
* laurence.calzone@curie.fr
Abstract
Understanding the etiology of metastasis is very important in clinical perspective, since it is
estimated that metastasis accounts for 90% of cancer patient mortality. Metastasis results
from a sequence of multiple steps including invasion and migration. The early stages of
metastasis are tightly controlled in normal cells and can be drastically affected by malignant
mutations; therefore, they might constitute the principal determinants of the overall meta-
static rate even if the later stages take long to occur. To elucidate the role of individual muta-
tions or their combinations affecting the metastatic development, a logical model has been
constructed that recapitulates published experimental results of known gene perturbations
on local invasion and migration processes, and predict the effect of not yet experimentally
assessed mutations. The model has been validated using experimental data on transcrip-
tome dynamics following TGF-β-dependent induction of Epithelial to Mesenchymal Transi-
tion in lung cancer cell lines. A method to associate gene expression profiles with different
stable state solutions of the logical model has been developed for that purpose. In addition,
we have systematically predicted alleviating (masking) and synergistic pairwise genetic
interactions between the genes composing the model with respect to the probability of
acquiring the metastatic phenotype. We focused on several unexpected synergistic genetic
interactions leading to theoretically very high metastasis probability. Among them, the syn-
ergistic combination of Notch overexpression and p53 deletion shows one of the strongest
effects, which is in agreement with a recent published experiment in a mouse model of gut
cancer. The mathematical model can recapitulate experimental mutations in both cell line
and mouse models. Furthermore, the model predicts new gene perturbations that affect the
early steps of metastasis underlying potential intervention points for innovative therapeutic
strategies in oncology.
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
1 / 29
a11111
OPEN ACCESS
Citation: Cohen DPA, Martignetti L, Robine S,
Barillot E, Zinovyev A, Calzone L (2015)
Mathematical Modelling of Molecular Pathways
Enabling Tumour Cell Invasion and Migration. PLoS
Comput Biol 11(11): e1004571. doi:10.1371/journal.
pcbi.1004571
Editor: Edwin Wang, National Research Council of
Canada, CANADA
Received: December 19, 2014
Accepted: September 29, 2015
Published: November 3, 2015
Copyright: © 2015 Cohen et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Funding: This work was funded by INVADE grant
from ITMO Cancer (Call Systems Biology 2012) to
DPAC LM AZ EB LC; and “Projet Incitatif et
Collaboratif Computational Systems Biology
Approach for Cancer” from Institut Curie; http://curie.
fr/recherche/programmes-incitatifs-cooperatifs-l%
E2%80%99institut-curie to DPAC LM AZ EB LC. The
funders had no role in study design, data collection
and analysis, decision to publish, or preparation of
the manuscript.
Author Summary
We provide here a logical model that proposes gene/pathway candidates that could abro-
gate metastasis. The model explores the mechanisms and interplays between pathways
that are involved in the process, identifies the main players in these mechanisms and gives
some insight on how the pathways could be altered. The model reproduces phenotypes of
published experimental results such as the double mutant Notch+/+/p53-/-. We have also
developed two methods: (1) to predict genetic interactions and (2) to match transcrip-
tomics data to the attractors of a logical model and validate the model on cell line
experiments.
Introduction
Understanding the etiology of metastasis is very important in clinical perspective. Despite the
progress with treatment of the primary tumours, the chances of survival for a patient decrease
tremendously once metastases have developed [1]. It is estimated that metastasis accounts for
90% of cancer patient mortality [2]. It is now understood that the metastatic process follows a
sequence of multiple steps, each being characterised by a small probability of success: 1) infil-
tration of tumour cells into the adjacent tissue, 2) migration of tumour cells towards vessels, 3)
intravasation of tumour cells by breaching through the endothelial monolayer, 4) travelling in
the circulatory or in the lymphoid system, 5) extravasation when circulating tumour cells re-
enter a distant tissue, and 6) colonisation and proliferation in distant organs [3]. The early
stages of invasion and migration are tightly controlled in normal cells and can be drastically
affected by malignant mutations. It has been shown indeed that primary and secondary
tumours have a common gene signature [4] that mediates the initial stages of metastasis while
extravasation and colony formation by a (tumour) cell does not require malignant gene alter-
ations [5], supporting the idea that the later stages of metastasis are affected by the anatomical
architecture of the vascular system [6].
Here, we focus on the ability of cancer cells to infiltrate and migrate into the surrounding
tissue. The first step towards the formation of secondary tumours is acquiring the ability to
migrate. In order to gain motile capacity, epithelial cells need to change their morphology
through Epithelial to Mesenchymal Transition (EMT), a process which occurs during develop-
ment (EMT type 1), wound healing (EMT type 2) and under pathological conditions such as
cancer (EMT type 3) [7,8]. EMT type 3 is characterised by both loss of E-cadherin (cdh1) and
invasive properties at the invasive front of the tumour [9]. Gene expression of E-cadherin is
inhibited by the transcription factors Snai1/2, Zeb1/2 and Twist1, while gene expression of N-
cadherin (cdh2) is induced by the same transcription factors [8,10,11]. These transcription
factors activate other genes that initiate EMT [11–13] and are induced by several signalling
pathways including TGF-β, NF-κB, Wnt and Notch pathways [8,14,15]. On the contrary, the
transcription factor p53 has been shown to inhibit EMT via degradation of Snai2 [16]; how-
ever, a p53 loss of function (LoF) alone is not sufficient to induce EMT [17]. After the switch of
E-cadherin to N-cadherin expression, the cell-cell contacts are weakened [18,19] and cancer
cells can pass the basal membrane to infiltrate the surrounding tissue [20]. The process of local
invasion can be active since tumour cells can secrete Matrix Metalloproteinases (MMPs) that
dissolve the lamina propria [21]. MMPs are also able to digest other components of the extra-
cellular matrix (ECM) and thereby to release growth factors and cytokines that are attached to
the ECM [21,22] which in turn activate the tumour cell’s ability to propagate the dissolvement
of the lamina propria. Finally, after dissolving the lamina propria and invading the (local)
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
2 / 29
Competing Interests: The authors have declared
that no competing interests exist.
stroma, cancer cells can migrate to distant sites by intravasation and extravasation of the vascu-
lar system [2]. To gain insight in the regulation of the metastatic process, several groups have
developed mathematical models of various aspects of it [23–29] (S1 Text).
Our aim is to understand the role of gene alterations in the development of metastasis. In
many (experimental or in silico) models, EMT is described as a very important step in acquir-
ing metastasis and even considered to be synonymous to appearance of metastasis [30–32].
Due to EMT role in metastasis, much research has been performed to elucidate its regulation.
The regulation of EMT is known to be complex and simple intuition is not sufficient to com-
prehend how genetic alterations (mutations and copy number variations) affect it. Logical
modelling can give qualitative insight on how they could affect EMT and subsequently
metastasis.
Previously, we have constructed a detailed map of molecular interactions involved in EMT
regulation which is freely available at [33], and based on its structural analysis, we hypothesized
a simple qualitative mechanism of EMT induction through upregulation of Notch and simulta-
neously deletion of p53. This prediction has been experimentally validated in a mouse model of
colon carcinoma [31].
In the present study, we significantly extend the biological context and provide a mathemat-
ical framework for the description of the necessary conditions for having metastasis, going
beyond the regulation of EMT only. We take into consideration the gained motility and ability
to invade as determinants of the metastatic process. For this purpose we largely extended and
re-designed the signalling network including more molecular players and phenotypes, and
translated the network into a formal mathematical model, allowing prediction of the metastasis
probability and the systematic analysis of mutant properties. Therefore, this work represents a
significant progress with respect to the previous results, allowing reconsideration of the qualita-
tive hypothesis suggested before using a formal mathematical modelling approach.
First, we introduce a logical model that recapitulates the molecular biology of the early steps
in metastasis. The construction of the influence network and the choice of the logical rules are
both based on knowledge derived from scientific articles. The final readouts of the model are
the phenotype variables CellCycleArrest, Apoptosis and the aggregated phenotype Metastasis
that combines the phenotypes EMT, Invasion and Migration.
We have chosen those final read-outs, as we believe that a metastatic phenotype depends on
the occurrence of EMT, invasion and migration. In addition, apoptosis is of importance to the
system as during healthy conditions, the cells undergo apoptosis when the cells detach from
the basal membrane [34]. Suppressing apoptosis during migration is a required key feature.
Our interest in cell cycle arrest is due to results of the mouse model [31] that show decreased
proliferation. We try to model this feature in our logical model by looking at the regulation of
cell cycle arrest. We did not focus on other phenotypes (or cancer hallmarks) such as prolifera-
tion explicitly, senescence, or angiogenesis. These are often considered in cancer studies but
they were out of the scope of this work, which focused on depicting early invasion modes and
not specifically on how tumour growth is regulated. The model inputs have been selected to
represent external signals necessary for the metastasis initiation pathways. The Boolean model
that we show here describes a possible regulation of the metastatic potential of a single tumour
cell and not of multiple cells or a tissue.
We provide a simplified version of the model where some genes are grouped into modules
(or pathways) allowing an analysis based on pathways rather than individual genes. Both ver-
sions of the models are validated by reproducing the phenotypic read-outs of published experi-
mental mouse and cell line models.
We then analyse the two models and formulate several types of predictions: at the level of
individual genes, e.g. exploring the individual role of each EMT inducer in metastasis; and at
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
3 / 29
the level of pathways, e.g. investigating the functional role of each pathway in triggering metas-
tasis. The logical models can suggest a systematic mechanistic explanation for the majority of
experimentally validated mutations on the local invasion and migration processes. Moreover,
we were able to establish a link between the solutions of the mathematical model and the gene
expression data from cell lines in which EMT was transiently induced. In addition, we have
applied this method to the analysis of transcriptomes of tumour biopsies.
Lastly, we investigate how genetic interactions between different gene mutations can affect
the probability of reaching a metastatic outcome. Our analysis predicts the effect of single
mutations and the genetic interactions between two single mutations with respect to several
cellular phenotypes. Our model proves an exceptionally efficient synergetic effect of increased
activity of Notch in combination with a decreased activity of p53 on metastasis in accordance
with our previous work [31].
Materials and Methods
The influence network
The construction of the influence network is based on scientific articles that describe the inter-
actions between nodes of the model. We first selected the main genes or proteins that may con-
tribute to activating EMT, regulating early invasion and triggering metastasis. We then
searched for experimentally proven physical interactions that would link all these players, and
simplified the detailed mechanisms into an influence network. For example, it has been shown
experimentally that AKT protein phosphorylates and stabilises MDM2, which in turn inhibits
p53 by forming a complex, leading to protein degradation of p53. We simplified the biochemi-
cal reactions by a negative influence from AKT to p53. The influence network is then translated
into a mathematical model using Boolean formalism (see below for details). We verified the
coherence of the network by comparing the outcome of the perturbed model to the observed
phenotypes of mutants found in the literature. The final model is the result of several iterations
that led to the accurate description of most of the published mutants related to the genes
included in our model. Once the model was able to reproduce most of the published mutant
experiments, we simulated mutants and conditions not yet assessed experimentally and pre-
dicted the outcome.
The Boolean formalism
From the influence network recapitulating known facts about the processes, we develop a math-
ematical model based on the Boolean formalism. To do so, we associate to a node of the influ-
ence network a corresponding Boolean variable. The variables can take two values: 0 for absent
or inactive (OFF), and 1 for present or active (ON). The variables change their value according
to a logical rule assigned to them. The state of a variable will thus depend on its logical rule,
which is based on logical statements, i.e., on a function of the node regulators linked with logi-
cal connectors AND (also denoted as &), OR (|) and NOT (!). A state of the model corresponds
to a vector of all variable states. All possible model states are connected into a transition graph
where the nodes are model states and the edges correspond to possible transitions from one
model state to another. The transition graph is based on asynchronous update, i.e., each vari-
able in the model state is updated one at a time as opposed to all together in the synchronous
update strategy. Attractors of the model refer to long-term asymptotic behaviours of the sys-
tem. Two types of attractors are identified: stable states, when the system has reached a model
state whose successor in the transition graph is itself; and cyclic attractors, when trajectories in
the transition graph lead to a group of model states that are cycling. In this model, no cyclic
attractors were found for the wild type case. However, we do not guarantee the non-existence
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
4 / 29
of cyclic attractors in some of the perturbed cases, as perturbations of the model may create
new dynamics.
The logical rules
A logical rule is written for each variable of the model, corresponding to a node in the influence
network, in order to define how its status evolves (ON or OFF). In this rule, the variables of the
input nodes are linked by logical connectors according to what is known about their combined
activities. There are several cases to consider: (1) The simplest logical rule that can be assigned
is when a variable depends on the activity of a single input: for instance, the transcription factor
Twist induces the transcription of the cdh2 gene (see Table 1); (2) In the case of an input that
has a negative effect on the activity of its target, the Boolean operator “NOT” or “!” is used:
EMT is, for example, activated by CDH2 but inactivated by CDH1, thus for EMT to be activate,
CDH1 should be OFF and CDH2 should be ON. The complete logical rule for the activation of
EMT will be EMT = 1 (ON) if CDH2 &! CDH1 (see Table 1); (3) In some cases, a gene can be
activated by two independent genes reflecting two different conditions and thus inputs are
Table 1. Logical rules describing the activity of a node.
Node
Rule
AKT1
CTNNB1 & (NICD | TGFbeta | GF | CDH2) & ! p53 & ! miR34 & ! CDH1
AKT2
TWIST1 & (TGFbeta | GF | CDH2) & !(miR203 | miR34 | p53)
CDH1
!TWIST1 & ! SNAI2 & ! ZEB1 & ! ZEB2 & ! SNAI1 & ! AKT2
CDH2
TWIST1
CTNNB1
!DKK1 & ! p53 & ! AKT1 & ! miR34 & ! miR200 & ! CDH1 & ! CDH2 & ! p63
DKK1
CTNNB1 | NICD
ERK
(SMAD | CDH2 | GF | NICD) & ! AKT1
GF
!CDH1 & (GF | CDH2)
miR200
(p63 | p53 | p73) & !(AKT2 | SNAI1 | SNAI2 | ZEB1 | ZEB2)
miR203
p53 & !(SNAI1 | ZEB1 | ZEB2)
miR34
!(SNAI1 | ZEB1 | ZEB2) & (p53 | p73) & AKT2 & ! p63 & ! AKT1
NICD
!p53 & ! p63 & ! p73 & ! miR200 & ! miR34 & ECM
p21
((SMAD & NICD) | p63 | p53 | p73 | AKT2) & !(AKT1 | ERK)
p53
(DNAdamage | CTNNB1 | NICD | miR34) & ! SNAI2 & ! p73 & ! AKT1 & ! AKT2
p63
DNAdamage & ! NICD & ! AKT1 & ! AKT2 & ! p53 & ! miR203
p73
DNAdamage & ! p53 & ! ZEB1 & ! AKT1 & ! AKT2
SMAD
TGFbeta & ! miR200 & ! miR203
SNAI1
(NICD | TWIST1) & ! miR203 & ! miR34 & ! p53 & ! CTNNB1
SNAI2
(TWIST1 | CTNNB1 | NICD) & ! miR200 & ! p53 & ! miR203
TGFbeta
(ECM | NICD) & ! CTNNB1
TWIST1
CTNNB1 | NICD | SNAI1
VIM
CTNNB1 | ZEB2
ZEB1
((TWIST1 & SNAI1) | CTNNB1 | SNAI2 | NICD) & ! miR200
ZEB2
(SNAI1 | (SNAI2 & TWIST1) | NICD) & ! miR200 & ! miR203
CellCycleArrest
(miR203 | miR200 | miR34 | ZEB2 | p21) & ! AKT1
Apoptosis
(p53 | p63 | p73 | miR200 | miR34) & ! ZEB2 & ! AKT1 & ! ERK
EMT
CDH2 & ! CDH1
Invasion
(SMAD & CDH2) | CTNNB1
Migration
VIM & AKT2 & ERK & ! miR200 & ! AKT1 & EMT & Invasion & ! p63
Metastasis
Migration
doi:10.1371/journal.pcbi.1004571.t001
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
5 / 29
linked by an OR, e.g., DKK can be activated either by CTNNB1 or by NICD, independently of
each other; (4) In some other cases, two activators are linked by an AND connector, e.g., ZEB2
whose activity depends on several inputs including TWIST1 & SNAI2 which are needed simul-
taneously: it has been observed experimentally that both transcription factors Twist1 and Snai2
are required to induce gene expression of zeb2. All models are available in GINsim format in
S1 File.
Computing phenotype probabilities using MaBoSS
MaBoSS is a C++ software for simulating continuous/discrete time Markov processes, defined
on the state transition graph describing the dynamics of a Boolean network. The rates up
(change from OFF to ON) and down (from ON to OFF) for each node are explicitly provided
in the MaBoSS configuration file together with logical functions, which allows working with
physical time explicitly. All rates are set to be 1 in this model since it is difficult to estimate
them from available experimental facts. Probabilities to reach a phenotype are computed as the
probability for the phenotype variable to have the value ON, by simulating random walks on
the probabilistic state transition graph. The probabilities for the selected outputs are reported
for each mutant based on predefined initial conditions (which can be all random). Since a state
in the state transition graph can combine the activation of several phenotype variables, not all
phenotype probabilities appear to be mutually exclusive. For example, Apoptosis phenotype
variable activation is always accompanied by activation of CellCycleArrest phenotype variable
(because p53 is a transcription factor of p21, responsible for cell cycle arrest, and the miRNAs,
activated by the p53 and its family members, lead to a cell cycle arrest), and activation of the
Metastasis phenotype variable is only possible when all three EMT, Invasion and Migration
phenotype variables are activated.
With MaBoSS, we can predict an increase or decrease of a phenotype probability when the
model variables are altered, which may correspond to the effect of particular mutants or drug
treatments. If mutation A increases the Apoptosis probability when compared to the Apoptosis
probability in wild type, we conclude that mutant A is advantageous for apoptosis. All models
are available in MaBoSS format in S1 File.
Module activity
The pathway activity (synonymously, module activity) score in a tumour sample is defined as
the contribution of this sample into the first principal component computed for all samples on
the set of the module target genes, as it was done in [35]. This way, we test target gene sets
selected from MSigDB [36] and KEGG [37] databases together with few tens of gene sets
assembled by us from external sources. The gene lists for each module is provided in S5 Table.
Differential activity score of each module was estimated by t-test between metastatic and non-
metastatic groups and significantly active/inactive modules were selected according to p-value
<0.05 condition.
Transcriptomics data for tumour samples
We conducted our study on the publicly available data of human colon cancer from TCGA
described in [38]. By excluding rectal cancers from the original dataset, the remaining 105
tumour samples were included in our analysis, classified into two groups (‘metastatic’ M1 = 17
tumours and ‘non-metastatic’ M0 = 88 tumours) according to clinical information about
metastasis appearance during cancer progression.
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
6 / 29
Transcriptomics data for cell lines
We used gene expression data generated from A549 lung adenocarcinoma cell line that was
treated with TGF-β1 ligand at eight different time points [39]. In short, gene expression was
measured for three replicates at each time point using Affymetrix Human Genome U133 Plus
2.0 Array. For more information about treatment and growth protocols see [40].
Matching transcriptomics data to logical steady states on EMT-induced
cancer cell lines
We followed the following six steps to link transcriptome data to the stable states of the model
(described in detail in S2 Text): (1) We first matched the genes of the model with their HUGO
names. For phenotypes such as Apoptosis, Migration or Invasion, the genes coding for CASP9,
CDC42, and MMP2 were used as biomarkers, respectively. These readouts were selected as the
most representative of the process; they were chosen based on the changes of the expression of
a list of candidate genes we explored throughout the experiments. (2) We averaged the genes
over the 3 replicates for time point T0 (initiation of experiment with no TGF-β), for T8 (identi-
fied as the beginning of EMT), for T24 (EMT in process) and for T72 (last point). (3) Using
several methods (binarization algorithms available in [41]), we identified a threshold of expres-
sion and binarized the data accordingly. Among our list of genes, only 11 of them have signifi-
cant expression dynamics along the experiment: cdh1, cdh2, ctnnb1, egfr, mapk1, mmp2,
smad3, snai2, tgfb1, vim and zeb1. The other genes were either always ON or always OFF
throughout the 72 hours of experiments because the expression is either above or below the
threshold we set. (4) We associated a label (phenotype) to the 9 stable states of the logical
model based on the activity status of the phenotype variable. (5) The similarity matrix was
computed according to the following rule: for each stable state and for each time point, if a
gene is ON (= 1) or OFF (= 0) in both the vector of discretized expression data and the vector
of the stable state, we set the entry in the similarity matrix to 1, otherwise, it is set to 0. (6) For
each time point and each stable state, we then summed up the corresponding similarity matrix
row, and set an expression-based phenotypic (EBP) score for each stable state. The highest EBP
score for each time point corresponds to the phenotype that is the closest to the studied sample
and is representative of the status of the cells.
Non-linear principal component analysis by elastic maps method
The non-linear principal manifold was constructed for the distribution of all single and double
mutants of the model in the space of computed model phenotype probabilities, using elastic
maps method and ViDaExpert software [42–44]. We preferred using a non-linear version of
principal component analysis (PCA) for data visualisation in this case, because it is known to
better preserve the local neighbourhood distance relations and allows more informative visual
estimation of clusters compared to the linear PCA of the same dimension [42]. For data analy-
sis, only those “mixed” phenotypes were selected whose probability expectation over the whole
set of single and double mutants was more than 1%. It resulted in a set of 1059 single and dou-
ble mutants embedded into 6-dimensional space of phenotype probabilities for which the prin-
cipal manifold was computed.
Synthetic interactions with respect to metastatic phenotype
The results of double mutants were used to quantify the level of epistasis between two model
gene defects (resulting either from gain-of-function mutation of a gene or from its knock-out
or loss-of-function mutation) with respect to metastatic phenotype. The level of epistasis was
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
7 / 29
quantified using the simplest multiplicative null model applied for the event of not having
metastasis: ε = (1-p12)-(1-p1)(1-p2), where p1 and p2 are the probabilities of having metastasis
in single mutants, and p12 was the probability of having metastasis in the double mutant.
Therefore, negative values of the epistasis score E correspond to synergistic interactions when
two gene defects amplify each other’s effect stronger than expected in the multiplicative model.
On the contrary, positive values correspond to alleviating effect, when the effect of one gene
defect could be masked (sometimes, even reduced to zero) by the second mutation. For genetic
network visualisation, we kept the most significant interactions with ε<-0.2 or ε>0.3 values.
These thresholds were chosen because at these levels we observed gaps in the distribution of ε
values. The complete list of interactions together with p1, p2, p12 and ε values can be found as a
Cytoscape 3 session (S2 File).
Results
Construction of an influence network regulating EMT, invasion and cell
migration
Mesenchymal cells are characterised by their increased motility, loss of cdh1 (coding for E-cad-
herin) expression, increased expression of cdh2 (coding for N-cadherin), and presence of
vimentin (Vim) [7,10,45]. The EMT program can be initiated by the transcription factors
snai1, snai2, zeb1, zeb2 and twist1. They are considered to be the core regulators of EMT as
each has been shown to down-regulate cdh1 [46–50]. In turn, the genes coding for these core
EMT-regulators are subjected to regulation by other signalling pathways. The TGF-β pathway
has been reported to be able to induce EMT [7,51], but other pathways are also involved in
EMT including Wnt, Notch and PI3K-AKT pathways [52–56].
Furthermore, microRNAs regulate the Snai and Zeb family members. For example, miR200
targets snai2, zeb1 and zeb2 mRNA [57–59] whereas miR203 targets snai1 and zeb2 mRNA
[59], and miR34 targets snai1 mRNA [60]. The transcription of these microRNAs is under the
control of p53 [61–64]. The miR200 expression can also be induced by p63 and p73 proteins,
while miR34 is only induced by p73 but is down-regulated by p63 [65–67]. The microRNAs
can be down-regulated by the EMT-inducers Snai1/2, and Zeb1/2 [59,60,68]. Note that the
proteins p63 and p73 have been identified as members of the p53 protein family since their
amino acid sequences share high similarity with that of p53 [69]. They are able to bind to the
promoters of the majority of the p53-target genes and therefore have overlapping functions in
cell cycle arrest and apoptosis [70,71]. The p53-family members are involved in cross-talks
with Notch and AKT pathways: p63 protein is inhibited by the Notch pathway, p53 by AKT1
and AKT2 [69,72–76] while p73 is down-regulated by p53 (itself negatively regulated by p73),
AKT1, AKT2, and Zeb1 [69,72,77].
The PI3K-AKT pathway has been considered to be important in evading apoptosis and cell
cycle arrest by modulating the TRAIL pathway, down-regulating pro-apoptotic genes and
phosphorylating p21 [78–80]. More recently, AKT has been assigned additional but important
roles in the development of metastasis. AKT1 suppresses apoptosis upon cell detachment
(anoikis) of the ECM [34]. The different isoforms of AKT seem to have opposing roles in the
regulation of microRNAs: AKT1 inhibits miR34 and activates miR200 while AKT2 inhibits
miR200 and activates miR34 [81]. Another opposing role for both AKT isoforms has been
found in migration. AKT1 inhibits migration by phosphorylating the protein Palladin; phos-
phorylated Palladin forms actin bundles that inhibit migration. AKT2 increases the protein
Palladin stability and upregulates β1-integrins stimulating migration [82,83] or by inhibiting
TSC2 that, in turn, activates RHO [84]. Furthermore, AKT1 inhibits cell cycle arrest while
AKT2 activates it [85,86] (all these effects are shown implicitly in Fig 1A).
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
8 / 29
Fig 1. Regulatory networks of mechanisms leading to EMT, invasion, migration and metastasis. A. Detailed network of the pathways involved in
metastasis. B. Modular network derived from network in A.
doi:10.1371/journal.pcbi.1004571.g001
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
9 / 29
Extracellular stimuli are also included in the logical model. Growth factors (GF) are soluble
ligands that can be excreted locally or from longer distances and are able to activate the
PI3K-AKT, and MAPK pathways [87,88]. Another extracellular stimulus might be the extracel-
lular microenvironment (ECMicroenv) with components that are not soluble including the
extracellular matrix. The ligands for the TGF-β pathway can be imbedded in the extracellular
matrix [89–91] and the ligands for the Notch pathway are transmembrane proteins from adja-
cent neighbouring cells [92,93].
These mechanisms are depicted in an influence network (Fig 1A). The network is composed
of nodes and edges, where some nodes represent biochemical species (proteins, miRNAs, pro-
cesses, etc.) and others represent phenotypes, and edges represent activating (green) or inhibi-
tory (red) influences of one node onto other node. Each edge is annotated and supported by
experimental papers (see S1 Table). Throughout the article, we will use the general term “phe-
notypes” to refer to “phenotype variables”, which correspond to the four outputs: CellCycleArr-
est, Apoptosis, Metastasis (depending on EMT, Migration and Invasion), and Homeostatic
State (HS) as presented below.
Mathematical modelling of the influence network
Construction of a logical model and its stable states. The network of Fig 1A is translated
into a logical model using GINsim software [94]: a logical rule is assigned to each node of the
network (Table 1, and Materials and Methods). Once the logical rules are set for each node of
the network, the Boolean model can simulate solutions or outcomes that correspond to attrac-
tors in the state transition graph (see Materials and Methods for details). The model, for the
wild type condition (i.e. no mutations or gene alterations), counts nine stable states for all com-
binations of inputs (Table 2). To each stable state, a phenotype is assigned based on the genes
that are activated (variable is ON, thus equal to 1). The phenotypes identified are: CellCycleArr-
est together with Apoptosis; CellCycleArrest together with EMT; Metastasis (depending on three
other processes: EMT, Migration and Invasion); and a stable state with only Cdh1 ON. This
state corresponds to a state where metastasis is inhibited by Cdh1 activity. We refer to it as the
homeostatic state (HS). It is a particular state of an epithelial cell that is not explicitly repre-
sented as a phenotype variable in this mathematical model. Four out of the nine stable states
lead to Apoptosis, in the presence of DNA damage and absence of growth factors (GF). Two sta-
ble states show an EMT phenotype alone (without inducing Metastasis). In these stable states,
Invasion and Migration are not activated because TGF-β pathway is not initially ON. The last
two stable states lead to Metastasis in the presence of growth factors. GF activates the ERK path-
way that switches off the p53-family targets and permits the triggering of events leading to
metastases. Indeed, several studies have shown the importance of ERK in migration [95–97].
Testing robustness of the model with respect to small changes in the logical rules.
We
systematically checked the effect of changing the logical operators of the model from “OR” to
“AND”, and vice versa, onto the resulting model phenotype probabilities. More specifically, we
generated model variants with one change of a logical operator in one logical rule, two changes
in the same logical rule, or one single change in two different logical rules, leaving the rest of
logical operators the same as in the wild type model. Therefore, we considered all model vari-
ants different from the wild type model by at most two different logical operators. The analysis
resulted in 8001 model variants.
We first show that the distributions of phenotype probabilities after these changes are con-
centrated around the wild type probability values (S6 Fig).
Metastasis appeared to be the least robust model phenotype, which confirms the fact that
there are some necessary conditions that need to be met to lead to metastasis (illustrated by
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
10 / 29
AND operators in the logical rules). If approximately 49% of changes in the logical rules have
minor or no effect onto the Metastasis phenotype probability, some modifications in some
rules changed the Metastasis phenotype to zero (implicating p63, p73, AKT1 variables of the
model and, to a lesser extent, CTNNB1, miR34, p53). Most of the rules that concern these vari-
ables are indeed more stringent. A change from an AND gate to an OR gate for the case of p63
or AKT1 has an important impact on the metastasis process. For instance, if p63 is more pres-
ent, because it is inactivated with fewer constraints, it can block more easily migration and
thus, metastasis. These logical rules should be considered more carefully than the others
because a mistake in defining these rules can have more drastic effects on the model properties
than any other modifications.
We also performed a reproducibility analysis of individual logical stable states in the models
differing from the wild type model by one or two changes in the same logical rule or one
change in two logical rules as presented above. The wild type model is characterized by nine
Table 2. The nine stable states of the mathematical model. The label of the columns corresponds to the phenotypic outputs.
HS
Apop1
Apop2
Apop3
Apop4
EMT1
EMT2
M1
M2
Metastasis
0
0
0
0
0
0
0
1
1
Migration
0
0
0
0
0
0
0
1
1
Invasion
0
0
0
0
0
0
0
1
1
EMT
0
0
0
0
0
1
1
1
1
Apoptosis
0
1
1
1
1
0
0
0
0
CellCycleArrest
0
1
1
1
1
1
1
1
1
ECMicroenv
0
0
0
1
1
0
0
1
1
DNAdamage
0
1
1
1
1
1
0
1
0
GF
0
0
0
0
0
1
1
1
1
TGFbeta
0
0
0
1
1
0
0
1
1
p21
0
1
1
1
1
0
0
0
0
CDH1
1
1
1
1
1
0
0
0
0
CDH2
0
0
0
0
0
1
1
1
1
VIM
0
0
0
0
0
1
1
1
1
TWIST1
0
0
0
0
0
1
1
1
1
SNAI1
0
0
0
0
0
1
1
1
1
SNAI2
0
0
0
0
0
1
1
1
1
ZEB1
0
0
0
0
0
1
1
1
1
ZEB2
0
0
0
0
0
1
1
1
1
AKT1
0
0
0
0
0
0
0
0
0
DKK1
0
0
0
0
0
0
0
1
1
CTNNB1
0
0
0
0
0
0
0
0
0
NICD
0
0
0
0
0
0
0
1
1
p63
0
0
1
0
1
0
0
0
0
p53
0
1
0
1
0
0
0
0
0
p73
0
0
1
0
1
0
0
0
0
miR200
0
1
1
1
1
0
0
0
0
miR203
0
1
0
1
0
0
0
0
0
miR34
0
0
0
0
0
0
0
0
0
AKT2
0
0
0
0
0
1
1
1
1
ERK
0
0
0
0
0
1
1
1
1
SMAD
0
0
0
0
0
0
0
1
1
doi:10.1371/journal.pcbi.1004571.t002
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
11 / 29
stable states, including the homeostatic state (HS) (see Table 2). 8001 model variants men-
tioned above are characterized by 68726 stable states counted in total. Hence, in average, each
model variant is characterized by 8 or 9 stable states, which might be different from the wild
type model. In total, we have counted 1176 distinct stable states in all the 8001 model variants,
observed with different frequencies (S6 Table). The nine stable states of the wild type model are
robustly reproducible, being the most frequently observed stable states, and accounting for
59% of all observed stable states in different model variants. Another 13% of observed stable
states differ from one of the wild type stable states by only one change in the Boolean variable
values (DIST_TO_WT = 1). Some modifications of logical rules for CTNNB1 or NICD lead to
very rarely observed atypical but very different from the wild type stable states (DIS-
T_TO_WT = 12). Based on all these analyses, we conclude that the nine wild type model stable
states are robust and “typical” with respect to moderate random modifications of the logical
rules and fragile to few targeted modifications.
Model reduction into a modular network.
To make our modelling more insightful, we
reduced the complexity by lumping variables into modules corresponding to signalling path-
ways: the TGF-β pathway (TGFb_pthw consisting of TGFbeta, SMAD), Notch pathway
(Notch_pthw, includes activated Notch intracellular domain (NICD), the WNT pathway
(WNT_pthw consisting of DKK1, CTNNB1), the p53 pathway (p53, consisting of p53), the
p63-p73 proteins (p63_73 consisting of p63 and p73), the miRNA (miR34, miR200, miR203),
the EMT regulators (EMT_reg including Twist1, Zeb1, Zeb2, Snai1, Snai2, Cdh2, Vim), E-
cadherin (Ecadh with Cdh1), growth factors (GF), the ERK pathway (ERK_pthw: ERK), p21 is
included in the CellCycleArrest phenotype, AKT1 module and AKT2 module. In the reduced
model (Fig 1B), the inputs (ECMicroenv and DNAdamage) and (final and intermediate) out-
puts (Migration, Invasion, Metastasis, and Apoptosis) are conserved. The reduced model pro-
duces the same stable states (for the wild type conditions) as those of the initial model (Fig 1A,
see S4 Text).
Validation of the Boolean model.
We simulated the genetic perturbations that corre-
spond to published experimental settings and verified that the stable states of the mathematical
model correspond to the experimental observations. An overexpression or gain of function
(GoF) of a gene is simulated by forcing the value of the node to ON and a deletion or loss of
function (LoF) by forcing the value of the node to OFF. We first simulated not only previously
described mutants but also mutants that have not yet been experimentally validated (see S4
Table). The mathematical model is able to reproduce the experimental results of almost all
described mutants. In few cases, there is a discrepancy between the mathematical and the bio-
logical model due to three reasons described below: 1) Metastasis in our logical model is
defined as colonisation of tumour cells into distant organs through migration in the systemic
and/or lymphatic vessels. A limitation of the cell line model is that a metastatic output cannot
be measured. 2) Dosage-dependent effects cannot be modelled using the logical formalism. For
example, our model predicts metastasis in a kras GoF while the mouse model does not develop
distant metastatic tumours. A possibility for the difference is that in the mouse model the wild
type kras allele is still present (a heterozygous mutation) while in our model KRAS mutant is
homozygous. It has been reported that the remaining wild type kras gene has still tumour sup-
pressive properties: it can reduce tumourigenesis in lung [98,99] and in colon cancer cell line
by inhibiting proliferation [100]. Other studies in cancer cell lines that are heterozygous for
kras mutation showed that the wild type kras gene in those cell lines decreased the migration
and colonisation capacity [101,102] suggesting a dose-dependent effect [103]. This might indi-
cate that mouse mutants homozygous for kras mutation may develop distant metastasis as pre-
dicted by our mathematical model. 3) Simulating mutations of genes that are not explicitly
represented as a node in the model has its limitations because the network does not describe
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
12 / 29
exactly the function of such node. For example, even though PTEN is not a variable of our
model, we simulated a pten mutation to understand the controversial results of such deletion
on metastasis in experimental models. The pten LoF mutations have been associated with
many different types of cancers [104–106] and recently it has been demonstrated that pten
mutations cause genomic instability [107,108]. In our model, in order to simulate a PTEN LoF,
its two targets, AKT1 and AKT2, are forced to be ON: PTEN inhibits activation of AKT iso-
forms [109–111]. The model predicts that a PTEN LoF alone or in combination with gene
mutations will reach the stable states without having metastasis while metastasis is observed in
the mouse model. In our model, due to activation of AKT1 by the PTEN LoF, metastasis is pre-
vented, because AKT1 inhibits migration as mentioned before. A recent study indicates that in
PTEN-deficient tumours, AKT2 is the active isoform [112] but not AKT1. The model confirms
this study: when we simulate the single AKT2 activation as a result of PTEN LoF, the model
predicts a stable state in which metastasis can be reached (All references and model results are
available in S4 Table).
Role of different pathways/modules in triggering metastasis
To assess the importance of each pathway on metastasis, apoptosis and cell cycle arrest, we
simulated a gain of function or a loss of function, in the reduced model, for each module and
for all combinations of inputs. These simulations mean that when an important entity in a
pathway is altered, it affects the whole pathway activity. The model shows that mutations lead-
ing to either GoF or LoF of each pathway have opposing results in the occurrence of migration
and for the occurrence of metastasis (S2 Table). The Notch_pthw is an exception in this: both a
GoF and LoF of the Notch pathway can lead to a stable state solution with metastasis ON. This
might indicate that Notch (pathway) activity must be in a certain range in order to have a non-
pathological effect or that Notch is important for the functioning of some dynamic feedback
controls preventing metastasis (so fixing it at a particular value would destroy these feedbacks).
In addition, GoF of the Notch_pthw, TGFb_pthw, ERK_pthw, EMT_reg or AKT2 shows their
inhibitory role in the apoptotic process as it has been demonstrated before [113–117]. For the
p53, TGF-β, EMT_reg and miRNA pathways, mutations leading to activation or inhibition
have opposing results in regulating invasion when either the pathway is activated or inhibited.
This effect on invasion is a direct result of having an activating or inhibiting role on EMT
except for the TGF-β pathway.
The role of TGF-β pathway has been investigated. The activation of TGF-β pathway might
be dependent on the micro-environment as its ligands can be found in the extracellular matrix
[89–91]. The triple mutant: Notch_pthw GoF, p53 LoF and TGFb_pthw LoF leads to one stable
state in which the EMT_reg is ON but no metastasis, migration, invasion or apoptosis are
reachable (S2 Table) indicating that activation of TGF-β pathway (e.g., by the peripheral
tumour cells more exposed to the micro-environment) is required to have metastasis in the
double mutant by activating invasion [118,119].
Comparing the Boolean model with dynamical transcriptomic data on EMT induction
and tumoral transcriptomes.
In this section, the aim is to investigate if the model can predict
temporal trends in the dynamics of high-throughput data in cancer cell lines or to retrospec-
tively predict a possible appearance of metastasis using the model. Is it possible to correlate
experimental or clinical data to the stable states of the model?
We first analysed the publicly available colon gene expression dataset generated by The
Cancer Genome Atlas (TCGA) project [38]. Student t-test between metastatic and non-
metastatic tumours was performed for genes included in the influence network to identify sig-
nificant changes in their expression between the two groups (S1 Fig). Few significant
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
13 / 29
differences were observed in the expression of the influence network genes in these two groups.
Moreover, there was no significant differential expression of the EMT regulators observed
between the two groups: expression of the EMT regulators seems to be OFF in these tumours.
Since single gene-based analysis of colon cancer did not show significant differential changes
in the expression of the influence network genes, we investigated the expression of the down-
stream targets for the transcription factors in the modular network (Notch_pthw, p53,
p63_p73, EMT regulators, etc.) and recapitulated their expression into a pathway activity score
(see Methods). The assumption was that the differential activity of a given transcription factor
can be better reflected by a score based on the expression of its target genes rather than from its
own individual expression. For the nodes that are not transcription factors (AKT1, AKT2, etc.)
we considered all genes involved in the same network module. We observed that the targets of
Notch pathway, Wnt pathway, p63_p73, and AKT1 and AKT2 downstream genes showed sig-
nificantly higher activity score in metastatic compared to non-metastatic samples whereas p53
and microRNAs targets were less active in metastatic samples. However, the EMT regulator
module showed almost no difference in module activity even if all regulators were combined in
one module (See S2 and S3 Figs). Indeed, in the recent colorectal tumour-specific EMT signa-
ture established by Tan et al. [120] none of the genes of our EMT module were included. This
means that at least in the colorectal cancer, the transcriptional dynamics of the EMT genes has
relatively small amplitude, when measured on the bulk of the tumour.
Based on our analysis, we hypothesize that only a small portion of the tumour cells in a
tumour sample are undergoing EMT and as a result, the EMT signal is strongly diluted when
looking at the whole cell population in a sample taken from the tumour bulk. This low signal to
noise ratio is not favourable to study the dynamics of the EMT process, and subsequently, the
metastatic process.
We thus analysed publicly available transcriptomics data from cancer cell lines in which
EMT has been induced. In a study conducted by Sartor and colleagues [39], lung carcinoma
cell lines were administered with increasing amount of TGF-β and genome-wide transcriptome
was measured at eight different time points, following the induction of EMT. The induction of
EMT was accompanied by increasing expression for some of the EMT regulators (S4 Fig).
The expression of these regulators follows a sigmoid curve in response to TGF-β induction.
For a given time-point, we checked if the expression level of the components of our model
could be associated to a particular steady state of the model. We expect our model to reflect the
behaviour of EMT expression level at early or late time points.
We then determined the consistency of the EMT induction experiment with the logical
model following the steps presented in Materials and Methods section (and in S2 Text for
details of each step). The resulting EBP (expression-based phenotypic) score of the method
represents how similar an experimental condition is to a stable state. Thus, the higher the EBP
score for a stable state is, the more similar the data are to that stable state, and as an extension,
to the phenotype variable associated to that stable state. The computed EBP scores at each time
point illustrate the evolution of the data in terms of the stable states. At T0, the highest EBP
score is associated with apoptosis. At T8, both metastasis and apoptosis stable states have the
highest EBP score illustrating the balance of phenotypes observed in the gradual entry into
EMT. At T24 and at T72, the metastatic phenotype has the highest EBP score suggesting that
EMT has occurred. Based on the above-mentioned results, the logical model is in accordance
with the time course experiments in EMT-induced cell lines.
With this similarity EBP score, we have developed a method to characterise tumours in
terms of a particular biological process (how the metastatic process follows EMT, migration
and invasion here) with respect to the solutions of a logical model.
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
14 / 29
Role of individual EMT regulators in triggering metastasis
To identify for each EMT regulator (Snai1, Snai2, Zeb1, Zeb2, Twist1) their specific role in the
different cell fates considered in our model, we simulated LoF and GoF mutants and observed
that all GoF, except for that of Snai2, led to the loss of apoptosis (S3 Table). Metastasis can be
reached for all GoF mutants but other phenotypes can still be reached depending on the combi-
nations of inputs. The single deletions of each EMT regulator show that Zeb2 and Twist1 are
required for metastasis. Zeb2 controls migration mainly through VIM but has no direct impact
on invasion. Twist1 LoF, on the contrary, modulates negatively the possibility to reach not only
the metastatic phenotype but also EMT, migration and invasion. Twist1 controls EMT through
Cdh2 that controls migration and EMT. Other factors, such as CTNNB1 (β-catenin) or TGF-β,
play a role in triggering the metastatic process by modulating invasion or migration, but our
model suggests that the main EMT regulators are Zeb2, Twist1 or Snai2, either as loss of func-
tion for Zeb2 and Twist1, or gain of function for Snai2. Note that by definition, Cdh2 is abso-
lutely required for metastasis to occur because of its direct role in controlling EMT and
migration. In our model, Cdh1 inhibits EMT (directly) and migration (through CTNNB1 and
VIM) but not invasion. Since all three phenotypes are required for metastasis, the process is
thus impaired when Cdh1 is over-expressed [121,122].
Modelling synthetic interactions between genes composing the model
The probability of achieving the metastatic phenotype for all possible single and double
mutants was systematically computed using MaBoSS [123]. Each single and double mutant is
characterised by the distribution of phenotype probabilities. A non-linear PCA analysis was
performed as described in Methods, which allowed to group together single and double
mutants having similar effect on the model phenotypes (Fig 2A). In this plot, one can
Fig 2. A. Genetic interactions between two mutants leading to the masking or the antagonism of a phenotype (metastasis). Application of non-linear
dimension reduction for visualising the distribution of phenotype probabilities, computed with MaBoSS for all single and double mutants of the model. The
grading in the background shows the density of points (mutants) projections. Six clusters are distinguished based on this grading. Wild-type model, all single
over-expression and knockout mutants and the NICD GoF / p53 LoF mutant are labelled. Note that each gene pair in this plot is represented by four different
double mutants (small red points) corresponding to LoF/LoF, LoF/GoF, GoF/LoF, GoF/GoF combinations. B. Genetic interaction network showing the most
significant synergistic (shown in green) and alleviating (masking, showing in red) interactions between GoF and LoF mutants with respect to the probability of
having metastasis. The size of the node reflects the metastasis probability for individual mutation. The thickness of the edge reflects the absolute value of
epistasis measure (see Methods).
doi:10.1371/journal.pcbi.1004571.g002
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
15 / 29
distinguish six major clusters (a to f) which can be tentatively annotated as “almost wild-type”
(no significant changes in the phenotype probabilities compared to the wild-type model), “risk
of metastasis” (elevated probability of having metastasis though not equal to 1), “apoptotic”
(for these mutants Apopotosis and CellCycleArrest phenotypes are activated), “EMT without
migration” (for these mutants, presented as two clusters, the formation of metastases cannot be
accomplished because the cells did not acquire ability to migrate), “cell cycle arrest only” (these
mutants are found arrested without starting EMT or invasion or apoptotic programs). The
direction of increased metastasis probability is shown by dashed line in Fig 2A, which ends at
NICD GoF/p53LoF double mutant for which the probability of having metastasis equals to 1,
according to the model (whereas single p53 LoF mutation belongs to “almost wild type” and
single NICD GoF mutation belongs to “risk of metastasis” clusters respectively).
Synthetic interactions with respect to metastatic phenotype. The most significant
genetic interactions with respect to probability of having metastasis (see Methods) are shown
in Fig 2B. The following observations can be made: (1) Hubs in this genetic interaction network
are the genes for which a single mutation (GoF) leads to a significant increase in having the
metastatic phenotype. These genes are akt2, twist1, snai1, and snai2; (2) There are a number of
genes whose LoF or GoF lead to a significantly masking effect on the phenotype caused by the
hub-gene mutations (red edges in Fig 2B). For example, overexpression of p53 gene or knock-
out of erk gene drastically decreases the probability of metastatic phenotype in SNAI1 LoF
mutant; (3) There are relatively few synergistic effects observed between single mutants (green
edges in Fig 2B). Some of them have been experimentally performed while other synergistic
interactions are rather unexpected such as GoF for both AKT2 and NICD, and can be a subject
of further experimental work.
There are four synergistic interactions, which result in augmenting the probability of having
metastasis to 100%. First, two of them are combinations of NICD GoF and p53 LoF (NICD+/
p53-), or simultaneous NICD GoF and p73 GoF (NICD+/p73+). These two interactions can be
considered as being dependent, since overexpression of p73 leads to downregulation of p53
function [124,125]. The other two interactions (SNAI2 GoF and NICD GoF, AKT2 GoF and
NICD GoF) are potential amplifier mechanisms for appearance of metastasis in NICD GoF
mutant alone.
In addition, we classified all gene pairs into five large clusters according to four different
combinations of in silico mutation types (LoF/LoF, LoF/GoF, GoF/LoF, GoF/GoF). Inside each
cluster, the gene pairs can be ranked according to the strength of the activating effect of one of
the mutation combinations on the Metastasis phenotype (S5 Fig). Moreover, all gene pairs can
be ranked according to the amplitude, i.e. the difference between the maximal and minimal
metastatic phenotype probabilities among four values (LoF/LoF, LoF/GoF, GoF/LoF, GoF/
GoF). The most distinguished gene pair in this analysis is p53/NICD, which is a unique and
extreme case of the gene pair cluster when any combination of mutation types besides LoF/
GoF makes the metastatic phenotype non-reachable (zero or close to zero probability) while
the synthetic-dosage interaction LoF/GoF makes the metastatic phenotype unavoidable (prob-
ability one) (cluster 3 in S5 Fig).
Synthetic dosage interaction between Notch and p53 genes.
Using MaBoSS, we have
been able to quantify the changes of probabilities for reaching each phenotype relative to the
wild type model. We are thus interested in results such as: “more or less apoptosis than in wild
type”. We simulated three mutants with MaBoSS: p53 LoF (Fig 3B), NICD GoF (Fig 3C) and
the double mutant NICD+/p53- (Fig 3D). These simulations are of particular interest since they
show an example of genetic interaction predicted to have the probability of metastasis pheno-
type equals to 1, as presented above.
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
16 / 29
The probabilities of the four phenotypes for wild type conditions are shown in Fig 3A. They
show all possible phenotypes for all input configurations. Note that Metastasis can be only
reached in the wild type for a particular set of initial conditions (ECMicroenv, GF and TGFbeta
all ON), which might correspond to extreme situations. The mutant p53 LoF is very similar to
the wild type in terms of possible phenotypes (Fig 2A). The NICD GoF mutation, compared to
the wild type, showed an increased probability for EMT as previously reported [126]. Metasta-
sis could be reached as well in this single mutant with a higher probability than in the wild
type; Apoptosis is no longer reachable, confirming that Notch pathway is a pro-survival path-
way (Figs 2A and 3C). In addition, in this mutant, Metastasis was clearly blocked by p53 since
a loss of function of p53 in a NICD GoF mutant completely suppressed both the EMT and
Fig 3. MaBoSS simulation of wild type, of individual mutations of p53 and NICD and of the double mutant. The probabilities associated with each
phenotype represent the number of stochastic simulations leading to each phenotype from pre-defined initial conditions. A. Wild type, see text. B. p53 LoF,
same phenotypes found in (A) are reachable but with different probabilities than wild type conditions. C. NICD GoF, apoptosis is no longer reachable. D. p53
LoF and NICD GoF, only metastasis is observed. Note that HS stands for “Homeostatic State” and CCA for “CellCycleArrest.”
doi:10.1371/journal.pcbi.1004571.g003
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
17 / 29
Invasion phenotypes present in the single mutant, and only Metastasis could be reached (Fig
3D). The deletion of both p63 and p73 in a NICD GOF mutant maintained the EMT pheno-
type (not shown) proving the importance of p53 in protecting the cells from metastasis.
In this context, we further investigated the role of TGF-β pathway in metastasis. Although
the NICD GoF/p53 LoF double mutation has been predicted to be the best mutation to acquire
metastasis, an important role for the TGF-β pathway is suggested by the model. The triple
mutant NICD GoF, p53 LoF and TGF-β LoF (S4 Table) seems to suppress the metastatic pro-
cess in the model: cells are able to go through EMT but cannot invade the tissue. Suppressing
the TGF-β pathway might be an interesting therapeutic option to control metastasis in patients;
however more studies are required to test this hypothesis.
Discussion
In this study, we propose a logical model focusing on the specific conditions that could allow
the occurrence of metastasis. Our model of the metastatic process represents its early steps:
EMT, invasion and migration. A cell acquires the capability to migrate when both EMT and
invasion abilities have been acquired. These steps are regulated by several signalling pathways,
where genetic aberrations could influence the efficiency of metastatic process. Both the influ-
ence network and the assignment of logical rules for each node of this network have been
derived from what has been published from experimental works as of today. With this model,
we were able to explore known conditions (and predict new ones) required for the occurrence
of metastasis. Our influence network describes the regulation of EMT, invasion, migration, cell
cycle arrest and apoptosis known from the literature. In this regulatory network, cell cycle
arrest and apoptosis are mechanisms or phenotypes that maintain homeostasis of organs [127]
or ways to evade metastasis. Cell migration depends on pathways involving AKT, ERK, Vimen-
tin, miR200 and p63 but also on the acquisition of EMT and invasive abilities such as produc-
ing MMPs to dissolve the laminae propria enabling migration to distant sites. Cells that have
only invasive properties are not able to move as they are still well attached to their surrounding
neighbouring cells resulting in absence of cell migration. Only when those two requirements
are met and the other pathways allow migration, can metastasis occur.
The role of each EMT regulator, for acquiring invasive properties, has been investigated and
the model shows that each individual EMT regulator is sufficient to induce EMT when over-
expressed and with the appropriate initial conditions. The model also predicts that a LoF muta-
tion of the EMT regulators does not affect metastasis except for ZEB2 and TWIST1: ZEB2 inhi-
bition leads to abrogation of migration, while a TWIST1 LoF leads to inhibition of EMT, since
TWIST1 is the only transcription factor that can induce transcription of cdh2 gene which is
required to have EMT. These regulators are interesting targets for therapy since both are more
downstream in the metastasis’ cascade knowing that most activating mutations occur relatively
more upstream e.g. KRAS and EGFR mutations.
The model has been validated using experimental data by matching the transcriptomic data
with stable state solutions of the logical model. The direct comparison between stable states
and gene expression of tumour samples shows no conclusive results. This may be due to that
only at the front of tumours, cells undergo EMT and this signal is obscured by the bulk of the
tumour [30,128]. On the other hand, the model matches well the transcriptomic data from a
time course experiment of lung carcinoma cell lines in which EMT was induced by increasing
concentration of TGF-β.
Qualitative simulations of the model using MaBoSS confirmed that single mutations are not
sufficient to enable metastasis. Therefore, we systematically computed the level of epistasis of
each two-gene mutation with respect to reaching the metastatic phenotype. We determined
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
18 / 29
which double mutations are the most efficient for inducing metastasis with NICD GoF/p53
LoF mutations being the most efficient combination of gene knock-out and over-dosage, as
this double mutant leads in silico to 100% probability of having metastasis.
In our previous work, this specific double mutation NICD GoF/p53 LoF has been carried
out experimentally in a mouse model, by crossing the villin-CreERT2 mice [129] (in this study
referred as p53 LoF) and RosaN1ic mice [130] (in this study referred as NICD GoF) with the
isogenic C57BL/6 animals to generate the NICD GoF/p53 LoF compound mice. These com-
pound mice develop intestinal tumours with metastatic tumours to distal organs [31]. Our logi-
cal model successfully reproduces experimental observations of the compound mouse and
proposes mechanisms explaining the metastatic phenotype with high penetrance in mice. In
addition, we have investigated the role of TGF-β pathway in metastasis and showed its crucial
role in the metastatic phenotype in the double mutant. Suppressing the TGF-β pathway might
be an interesting target therapy to control metastasis, however future studies are required.
We also explored the activity of the Wnt pathway in the double mutant. Increased activity
of the Wnt pathway due to mutations in the apc and ctnnb1 genes leads to tumourigenesis of
many cancers [131–133] and subsequently to metastasis [134,135]. Our mathematical model
predicts phenotypes that correspond to adenocarcinomas as a result of linear progression of
acquired mutations during sporadic colorectal cancer (CRC) suggested by the “Vogelstein
sequence” [136] but no metastasis is reached with the model. Indeed, when we simulate APC
LoF, KRAS GoF and p53 LoF (the Vogelstein sequence), the model predicts stable states of
cells that are not arrested in the cell cycle, can undergo EMT and can invade (see S4 Table).
Thus our logical model supports the hypothesis that the Wnt pathway contributes to tumour
initiation [137]. However, there is still a debate if the Wnt pathway is actively involved in
metastasis. For example, a negative correlation has been demonstrated between the presence of
β-catenin and metastasis in breast cancer [138], in lung cancer [139–141], and in CRC [142–
144]. It has been also demonstrated that the canonical Wnt pathway (β-catenin-dependent
pathway) is suppressed at the leading edge of the tumour [145] and this might happen without
affecting the β-catenin protein levels [146,147]. In the mouse model with Notch GoF /p53 LoF
double mutation, in some tumours samples, mutations in apc and ctnnb1 have been found but
also tumours without those mutations have been shown to acquire metastasis. Both truncated
APC and mutations in β-catenin correspond in our mathematical model to full activation of
CTNNB1 and this will induce activation of AKT1. In our model, activation of AKT1 will
inhibit migration and therefore inhibit metastasis. Appearance of metastasis in the mouse
model with activated Wnt pathway might be putatively explained if one looks at the length of
the truncated APC isoform for tumours with apc mutation. The APC mutation found in the
Notch GoF /p53 LoF mouse model results in a relatively large truncated APC isoform that
might still have inhibitory effect on β-catenin [148]. More details about the APC isoforms and
its effect on β-catenin can be found in S3 Text.
Another explanation for having metastasis in tumours with active Wnt pathway might be
the involvement of another mutation that affects the akt1 or the akt2 gene. According to our
model, the Wnt pathway inhibits metastasis by up-regulation of AKT1. There are tumours in
CRC patients (TCGA data from http://cbioportal.org, [31]) that can have an akt2 gene amplifi-
cation or a homozygous deletion or missense mutation of akt1. AKT2 induces migration while
AKT1 inhibits migration thus the ratio AKT1 to AKT2 might be an important determinant for
acquiring metastasis in the colon. Indeed studies have shown that AKT2 is predominant in
sporadic colon cancer [149] and have a critical role in metastasis in CRC [150].
A Boolean model of EMT induction has been recently published, where the theoretical pre-
diction that the Wnt pathway can be activated upon TGF-β administration was validated
experimentally by measuring increased gene expression of the Wnt target gene axin2 in Huh7
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
19 / 29
and PLC/PRF/5 cell lines [151]. Those cell lines are derived from hepatocellular carcinomas
[152,153] and both can harbour known mutations [154] and unconfirmed mutations (http://
tinyurl.com/l6mjd8y) that affect the signalling pathways: the Wnt pathway has constitutive
activity in the Huh7 cell line [137,155]. An alternative explanation could be that our model is
more specific for epithelial cancers as the model depicts many reactions observed in epithelial
cells; it has been shown that different types of cancer have different protein (or isoforms) abun-
dance [112,149]. Therefore, our model might be less adequate in predicting the activity for cer-
tain nodes for hepatocellular carcinoma and lung adenocarcinoma.
EMT is considered to be the first step and is very often modelled as an equivalent of having
metastasis once it is activated. We provide here a logical model that proposes the involvement
of three independent processes in order to have metastasis: EMT, invasion and migration.
These phenotypes are controlled by an intricate network and only when EMT, invasion and
migration do occur, can metastasis happen. The logical model explores the mechanisms and
interplays between pathways that are involved in the processes, identifies the main players in
these mechanisms and gives insight on how these pathways could be altered in a therapeutic
perspective. Note that other mechanisms involving other alterations in the pathways that we
model, or in other pathways might also take place, and we do not claim that our approach
cover all possibilities of inducing metastasis. Still, our approach provides candidate interven-
tion points for designing innovative anti-metastatic strategies.
Supporting Information
S1 Text. Review on published articles of mathematical models of EMT.
(DOCX)
S2 Text. Link between model solutions and transcriptomics data.
(DOCX)
S3 Text. Description of Wnt pathway.
(PDF)
S4 Text. From the master model to the reduced model.
(DOCX)
S1 Fig. Colon transcriptomics data. Mean value expression for each gene is mapped on the
network. The figure is the same for both metastatic and non-metastatic samples.
(PNG)
S2 Fig. Modular network of the metastasis model.
(PDF)
S3 Fig. Colon transcriptomics data mapped onto the modular network. The score for the
modules are calculated based on the expression of target genes for metastatic and non-
metastatic samples.
(PDF)
S4 Fig. Mean gene expression value of the three replicates for the genes of the network at 4
different time points: At t = 0, at t = 8h, at t = 24h and at t = 72h. Green nodes correspond to
low expression and red nodes to high expression. The minimum and maximum expression val-
ues are set over the whole dataset and are the same for the four graphs.
(PDF)
S5 Fig. Distribution of pairs of genes of the mathematical model in the four-dimensional
space of Metastasis probabilities, corresponding to four possible mutation type
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
20 / 29
combinations LoF/LoF, LoF/GoF, GoF/LoF, GoF/GoF (here LoF is Loss-of-Function and
GoF is Gain-of-Function). The image shows a two-dimensional projection onto a non-linear
principal manifold from the space defined by four metastatic phenotype probabilities [p(LoF/
LoF);p(GoF/GoF); p(LoF/GoF)+p(GoF/LoF);|p(LoF/GoF)-p(GoF/LoF)|]. Projection density is
shown in the background by grey shading. The size of the node corresponds to the amplitude
of the node pair (maximum difference in phenotype probability between the four mutants:
LoF/LoF, GoF/GoF, GoF/LoF, LoF/GoF), such that the most sensitive (allowing control of phe-
notype to maximal degree) gene pairs correspond to bigger node sizes. Five clusters are identi-
fied: they correspond to five patterns which existence can be guessed from the symmetry
considerations and which are shown on the right panels. 1a) Any GoF cancels the phenotype
while double LoF can amplify it (14% of gene pairs); 1b) Any LoF cancels the phenotype while
double GoF can amplify it (13%); 2a) Double GoF cancels the phenotype, double LoF or syn-
thetic-dosage interaction can amplify it (23%); 2b) Double LoF cancels the phenotype, double
GoF or synthetic-dosage interaction (LoF/GoF or GoF/LoF) can amplify it (16%); 3) Double
LoF and double GoF cancel the phenotype, while synthetic-dosage interaction can amplify it
(30%). TP53-NICD (top-left corner) mutant is an extreme example of group 3. NICD-AKT2
(bottom-left corner) is an extreme example of group 2b.
(PNG)
S6 Fig. Results of robustness tests for the logical model with respect to small changes in the
logical rules of the model. In the wild type logical model, for each logical rule, several "variant"
models were created by changing one or two "OR" or "AND" operators to "AND" or "OR" oper-
ators respectively. The resulting distributions of phenotype probabilities over all such model
modifications are shown.
(PDF)
S1 Table. Annotations of the logical model.
(DOCX)
S2 Table. Phenotypes that can be reached by setting the activity of a single module or path-
way to always ON (GoF: gain of function) or always OFF (LoF: loss of function).
(XLSX)
S3 Table. Phenotypes that can be reached by setting the activity of a single EMT regulator
to always ON (GoF: gain of function) or always OFF (LoF: loss of function). EMT regula-
tors: Snai1, Snai2, Zeb1, Zeb2, and Twist1.
(XLSX)
S4 Table. Table of mutants. For each condition or mutation, all possible inputs are considered.
Thus, all possible outputs corresponding to stable states are shown in this table (values for
internal variables are not shown). The existence of a stable state in accordance with what has
been published is enough to conclude that the mutant is validated: there exists a condition
for which the model explains the experiments. The fact that other stable states exist shows
that for some particular conditions, the stable state could be reachable. For instance, for NICD
GoF, we see that a stable state with metastasis exits which has not been observed in experi-
ments. However, for this stable state, all p53 family members are OFF, thus, it is a particular sit-
uation.
(DOCX)
S5 Table. Signatures of module activity.
(XLS)
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
21 / 29
S6 Table. Robustness analysis; table of mutants for the logical stable states of the perturbed
models.
(XLSX)
S1 File. Detailed and modular models in GINsim and MaBoSS formats. The zip file
includes: Detailed model in GINsim format (SuppMat_Model_Master_Model.zginml), Modu-
lar model in GINsim format (SuppMat_Model_ModNet.zginml), SuppMat_MaBoSS_Master-
Model.bnd (To simulate the model, MaBoSS needs to be downloaded from maboss.curie.fr and
launched with the following command line:./MaBoSS -c SuppMat_MaBoSS_MasterModel.cfg
-o SuppMat_MaBoSS_MasterModel SuppMat_MaBoSS_MasterModel.bnd), SuppMat_Ma-
BoSS_MasterModel.cfg, SuppMat_MaBoSS_ModNet.bnd (To simulate the model, MaBoSS
needs to be downloaded from maboss.curie.fr and launched with the following command
line:./MaBoSS -c SuppMat_MaBoSS_ModNet.cfg -o SuppMat_MaBoSS_ModNet SuppMat_
MaBoSS_ModNet.bnd), SuppMat_MaBoSS_ModNet.cfg.
(ZIP)
S2 File. SuppMat_metastasis_mutants.cys.
(ZIP)
Acknowledgments
We are thankful to Prof Dr Daniel Louvard for critical reading and advising on the
manuscript.
Author Contributions
Conceived and designed the experiments: DPAC LM SR EB AZ LC. Performed the experi-
ments: DPAC LM AZ LC. Analyzed the data: LM DPAC AZ LC. Contributed reagents/
materials/analysis tools: DPAC LM AZ LC. Wrote the paper: DPAC LM AZ EB LC. Contrib-
uted to the model construction: DPAC SR EB AZ LC. Corrected the manuscript: SR EB.
References
1.
Siegel R, Ward E, Brawley O, Jemal A Cancer statistics, 2011: the impact of eliminating socioeco-
nomic and racial disparities on premature cancer deaths. CA Cancer J Clin 61: 212–236. doi: 10.
3322/caac.20121 PMID: 21685461
2.
Reymond N, D’Água BB, Ridley AJ (2013) Crossing the endothelial barrier during metastasis. Nat
Rev Cancer 13: 858–870. doi: 10.1038/nrc3628 PMID: 24263189
3.
Van Zijl F, Krupitza G, Mikulits W (2011) Initial steps of metastasis: cell invasion and endothelial trans-
migration. Mutat Res 728: 23–34. doi: 10.1016/j.mrrev.2011.05.002 PMID: 21605699
4.
Ramaswamy S, Ross KN, Lander ES, Golub TR (2003) A molecular signature of metastasis in pri-
mary solid tumors. Nat Genet 33: 49–54. doi: 10.1038/ng1060 PMID: 12469122
5.
Podsypanina K, Du Y-CN, Jechlinger M, Beverly LJ, Hambardzumyan D, et al. (2008) Seeding and
propagation of untransformed mouse mammary cells in the lung. Science 321: 1841–1844. doi: 10.
1126/science.1161621 PMID: 18755941
6.
Conteduca V, Zamarchi R, Rossi E, Condelli V, Troiani L, et al. (2013) Circulating tumor cells: utopia
or reality? Future Oncol 9: 1337–1352. doi: 10.2217/fon.13.101 PMID: 23980681
7.
Thiery JP, Acloque H, Huang RYJ, Nieto MA (2009) Epithelial-mesenchymal transitions in develop-
ment and disease. Cell 139: 871–890. doi: 10.1016/j.cell.2009.11.007 PMID: 19945376
8.
Foroni C, Broggini M, Generali D, Damia G (2012) Epithelial-mesenchymal transition and breast can-
cer: Role, molecular mechanisms and clinical impact. Cancer Treat Rev 38: 689–697. doi: 10.1016/j.
ctrv.2011.11.001 PMID: 22118888
9.
Kalluri R, Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J Clin Invest 119:
1420–1428. doi: 10.1172/JCI39104 PMID: 19487818
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
22 / 29
10.
Zeisberg M, Neilson EG (2009) Biomarkers for epithelial-mesenchymal transitions. J Clin Invest 119:
1429–1437. doi: 10.1172/JCI36183 PMID: 19487819
11.
Sánchez-Tilló E, Liu Y, de Barrios O, Siles L, Fanlo L, et al. (2012) EMT-activating transcription factors
in cancer: beyond EMT and tumor invasiveness. Cell Mol Life Sci 69: 3429–3456. doi: 10.1007/
s00018-012-1122-2 PMID: 22945800
12.
Nieto MA (2011) The ins and outs of the epithelial to mesenchymal transition in health and disease.
Annu Rev Cell Dev Biol 27: 347–376. doi: 10.1146/annurev-cellbio-092910-154036 PMID: 21740232
13.
Geiger TR, Peeper DS (2009) Metastasis mechanisms. Biochim Biophys Acta 1796: 293–308. doi:
10.1016/j.bbcan.2009.07.006 PMID: 19683560
14.
Polyak K, Weinberg RA (2009) Transitions between epithelial and mesenchymal states: acquisition of
malignant and stem cell traits. Nat Rev Cancer 9: 265–273. doi: 10.1038/nrc2620 PMID: 19262571
15.
Thiery JP, Sleeman JP (2006) Complex networks orchestrate epithelial-mesenchymal transitions. Nat
Rev Mol Cell Biol 7: 131–142. doi: 10.1038/nrm1835 PMID: 16493418
16.
Wang S-P, Wang W-L, Chang Y-L, Wu C-T, Chao Y-C, et al. (2009) p53 controls cancer cell invasion
by inducing the MDM2-mediated degradation of Slug. Nat Cell Biol 11: 694–704. doi: 10.1038/
ncb1875 PMID: 19448627
17.
Jonkers J, Meuwissen R, van der Gulden H, Peterse H, van der Valk M, et al. (2001) Synergistic
tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nat
Genet 29: 418–425. doi: 10.1038/ng747 PMID: 11694875
18.
Wijnhoven BP, Dinjens WN, Pignatelli M (2000) E-cadherin-catenin cell-cell adhesion complex and
human cancer. Br J Surg 87: 992–1005. doi: 10.1046/j.1365-2168.2000.01513.x PMID: 10931041
19.
Le Bras GF, Taubenslag KJ, Andl CD (2012) The regulation of cell-cell adhesion during epithelial-
mesenchymal transition, motility and tumor progression. Cell Adh Migr 6: 365–373. doi: 10.4161/
cam.21326 PMID: 22796940
20.
Bravo-Cordero JJ, Hodgson L, Condeelis J (2012) Directed cell invasion and migration during metas-
tasis. Curr Opin Cell Biol 24: 277–283. doi: 10.1016/j.ceb.2011.12.004 PMID: 22209238
21.
Wan L, Pantel K, Kang Y (2013) Tumor metastasis: moving new biological insights into the clinic. Nat
Med 19: 1450–1464. doi: 10.1038/nm.3391 PMID: 24202397
22.
Bauvois B (2012) New facets of matrix metalloproteinases MMP-2 and MMP-9 as cell surface trans-
ducers: outside-in signaling and relationship to tumor progression. Biochim Biophys Acta 1825:
29–36. doi: 10.1016/j.bbcan.2011.10.001 PMID: 22020293
23.
Barillot E, Calzone L, Hupé P, Vert J-P, Zinovyev A (2012) Computational Systems Biology of Cancer.
Chapman & Hall/CRC. 461 p.
24.
Anderson ARA, Weaver AM, Cummings PT, Quaranta V (2006) Tumor morphology and phenotypic
evolution driven by selective pressure from the microenvironment. Cell 127: 905–915. doi: 10.1016/j.
cell.2006.09.042 PMID: 17129778
25.
Martin NK, Gaffney EA, Gatenby RA, Maini PK (2010) Tumour-stromal interactions in acid-mediated
invasion: a mathematical model. J Theor Biol 267: 461–470. doi: 10.1016/j.jtbi.2010.08.028 PMID:
20816684
26.
Knutsdottir H, Palsson E, Edelstein-Keshet L (2014) Mathematical model of macrophage-facilitated
breast cancer cells invasion. J Theor Biol. doi: 10.1016/j.jtbi.2014.04.031
27.
Lu M, Jolly MK, Levine H, Onuchic JN, Ben-Jacob E (2013) MicroRNA-based regulation of epithelial-
hybrid-mesenchymal fate determination. Proc Natl Acad Sci U S A 110: 18144–18149. doi: 10.1073/
pnas.1318192110 PMID: 24154725
28.
Tian X-J, Zhang H, Xing J (2013) Coupled reversible and irreversible bistable switches underlying
TGFβ-induced epithelial to mesenchymal transition. Biophys J 105: 1079–1089. doi: 10.1016/j.bpj.
2013.07.011 PMID: 23972859
29.
Zhang J, Tian X-J, Zhang H, Teng Y, Li R, et al. (2014) TGF-β-induced epithelial-to-mesenchymal
transition proceeds through stepwise activation of multiple feedback loops. Sci Signal 7: ra91. doi: 10.
1126/scisignal.2005304
30.
Sethi S, Macoska J, Chen W, Sarkar FH (2010) Molecular signature of epithelial-mesenchymal transi-
tion (EMT) in human prostate cancer bone metastasis. Am J Transl Res 3: 90–99. PMID: 21139809
31.
Chanrion M, Kuperstein I, Barrière C, El Marjou F, Cohen D, et al. (2014) Concomitant Notch activa-
tion and p53 deletion trigger epithelial-to-mesenchymal transition and metastasis in mouse gut. Nat
Commun 5: 5005. doi: 10.1038/ncomms6005 PMID: 25295490
32.
Tsai JH, Yang J (2013) Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev 27:
2192–2206. doi: 10.1101/gad.225334.113 PMID: 24142872
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
23 / 29
33.
Signaling network of EMT regulation http://navicell.curie.fr/navicell/maps/signallingnetworkofemtregulation/
master/index.html
34.
Kim Y-N, Koo KH, Sung JY, Yun U-J, Kim H (2012) Anoikis resistance: an essential prerequisite for
tumor metastasis. Int J Cell Biol 2012: 306879. doi: 10.1155/2012/306879 PMID: 22505926
35.
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, et al. (2005) Gene set enrichment
analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl
Acad Sci U S A 102: 15545–15550. doi: 10.1073/pnas.0506580102 PMID: 16199517
36.
Bild AH, Yao G, Chang JT, Wang Q, Potti A, et al. (2006) Oncogenic pathway signatures in human
cancers as a guide to targeted therapies. Nature 439: 353–357. doi: 10.1038/nature04296 PMID:
16273092
37.
Kanehisa M, Goto S, Kawashima S, Okuno Y, Hattori M (2004) The KEGG resource for deciphering
the genome. Nucleic Acids Res 32: D277–D280. doi: 10.1093/nar/gkh063 PMID: 14681412
38.
The Henome Cancer Atlas Network (2012) Comprehensive molecular characterization of human
colon and rectal cancer. Nature 487: 330–337. doi: 10.1038/nature11252 PMID: 22810696
39.
Sartor MA, Mahavisno V, Keshamouni VG, Cavalcoli J, Wright Z, et al. (2010) ConceptGen: a gene
set enrichment and gene set relation mapping tool. Bioinformatics 26: 456–463. doi: 10.1093/
bioinformatics/btp683 PMID: 20007254
40.
Qin H, Chan MWY, Liyanarachchi S, Balch C, Potter D, et al. (2009) An integrative ChIP-chip and
gene expression profiling to model SMAD regulatory modules. BMC Syst Biol 3: 73. doi: 10.1186/
1752-0509-3-73 PMID: 19615063
41.
Müssel C, Hopfensitz M, Kestler HA (2010) BoolNet—an R package for generation, reconstruction
and analysis of Boolean networks. Bioinformatics 26: 1378–1380. doi: 10.1093/bioinformatics/btq124
PMID: 20378558
42.
Gorban A, Kegl B, Wunch D, Zinovyev A, editors (2008) Principal Manifolds for Data Visualisation and
Dimension Reduction. Vol. 58. Berlin-Heidelberg: Springer. 340 p.
43.
Gorban AN, Zinovyev A (2010) Principal manifolds and graphs in practice: from molecular biology to
dynamical systems. Int J Neural Syst 20: 219–232. doi: 10.1142/S0129065710002383 PMID:
20556849
44.
Gorban A., Pitenko A., Zinovyev A (2014) ViDaExpert: user-friendly tool for nonlinear visualization
and analysis of multidimensional vectorial data. URL: http://arxiv.org/abs/1406.5550
45.
Drasin DJ, Robin TP, Ford HL (2011) Breast cancer epithelial-to-mesenchymal transition: examining
the functional consequences of plasticity. Breast Cancer Res 13: 226. doi: 10.1186/bcr3037 PMID:
22078097
46.
Moreno-Bueno G, Cubillo E, Sarrió D, Peinado H, Rodríguez-Pinilla SM, et al. (2006) Genetic profiling
of epithelial cells expressing E-cadherin repressors reveals a distinct role for Snail, Slug, and E47 fac-
tors in epithelial-mesenchymal transition. Cancer Res 66: 9543–9556. doi: 10.1158/0008-5472.CAN-
06-0479 PMID: 17018611
47.
Conacci-Sorrell M, Simcha I, Ben-Yedidia T, Blechman J, Savagner P, et al. (2003) Autoregulation of
E-cadherin expression by cadherin-cadherin interactions: the roles of beta-catenin signaling, Slug,
and MAPK. J Cell Biol 163: 847–857. doi: 10.1083/jcb.200308162 PMID: 14623871
48.
Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, et al. (2004) Twist, a master regulator of
morphogenesis, plays an essential role in tumor metastasis. Cell 117: 927–939. doi: 10.1016/j.cell.
2004.06.006 PMID: 15210113
49.
Liu Y, El-Naggar S, Darling DS, Higashi Y, Dean DC (2008) Zeb1 links epithelial-mesenchymal transi-
tion and cellular senescence. Development 135: 579–588. doi: 10.1242/dev.007047 PMID:
18192284
50.
Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, et al. (2001) The two-handed E
box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell 7:
1267–1278. PMID: 11430829
51.
Katsuno Y, Lamouille S, Derynck R (2013) TGF-β signaling and epithelial-mesenchymal transition in
cancer progression. Curr Opin Oncol 25: 76–84. PMID: 23197193
52.
Mao Y, Xu J, Li Z, Zhang N, Yin H, et al. (2013) The role of nuclear β-catenin accumulation in the
Twist2-induced ovarian cancer EMT. PLoS One 8: e78200. doi: 10.1371/journal.pone.0078200
PMID: 24244294
53.
Espinoza I, Miele L (2013) Deadly crosstalk: Notch signaling at the intersection of EMT and cancer
stem cells. Cancer Lett. doi: 10.1016/j.canlet.2013.08.027
54.
Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, et al. (2004) Notch promotes
epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes
Dev 18: 99–115. doi: 10.1101/gad.276304 PMID: 14701881
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
24 / 29
55.
Grille SJ, Bellacosa A, Upson J, Klein-Szanto AJ, van Roy F, et al. (2003) The protein kinase Akt
induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squa-
mous cell carcinoma lines. Cancer Res 63: 2172–2178. PMID: 12727836
56.
Suman S, Kurisetty V, Das TP, Vadodkar A, Ramos G, et al. (2013) Activation of AKT signaling pro-
motes epithelial-mesenchymal transition and tumor growth in colorectal cancer cells. Mol Carcinog.
doi: 10.1002/mc.22076
57.
Hill L, Browne G, Tulchinsky E (2012) ZEB/miR-200 feedback loop: At the crossroads of signal trans-
duction in cancer. Int J Cancer. doi: 10.1002/ijc.27708
58.
Liu Y-N, Yin JJ, Abou-Kheir W, Hynes PG, Casey OM, et al. (2012) MiR-1 and miR-200 inhibit EMT
via Slug-dependent and tumorigenesis via Slug-independent mechanisms. Oncogene. doi: 10.1038/
onc.2012.58
59.
Moes M, Le Béchec A, Crespo I, Laurini C, Halavatyi A, et al. (2012) A Novel Network Integrating a
miRNA-203/SNAI1 Feedback Loop which Regulates Epithelial to Mesenchymal Transition. PLoS
One 7: e35440. doi: 10.1371/journal.pone.0035440 PMID: 22514743
60.
Siemens H, Jackstadt R, Hünten S, Kaller M, Menssen A, et al. (2011) miR-34 and SNAIL form a dou-
ble-negative feedback loop to regulate epithelial-mesenchymal transitions. Cell Cycle 10: 4256–
4271. doi: 10.4161/cc.10.24.18552 PMID: 22134354
61.
Hao L, Ha JR, Kuzel P, Garcia E, Persad S (2012) Cadherin switch from E- to N-cadherin in mela-
noma progression is regulated by the PI3K/PTEN pathway through Twist and Snail. Br J Dermatol
166: 1184–1197. doi: 10.1111/j.1365-2133.2012.10824.x PMID: 22332917
62.
McKenna DJ, McDade SS, Patel D, McCance DJ (2010) MicroRNA 203 expression in keratinocytes
is dependent on regulation of p53 levels by E6. J Virol 84: 10644–10652. doi: 10.1128/JVI.00703-10
PMID: 20702634
63.
Tarasov V, Jung P, Verdoodt B, Lodygin D, Epanchintsev A, et al. (2007) Differential regulation of
microRNAs by p53 revealed by massively parallel sequencing: miR-34a is a p53 target that induces
apoptosis and G1-arrest. Cell Cycle 6: 1586–1593. PMID: 17554199
64.
Kim NH, Kim HS, Li X-Y, Lee I, Choi H-S, et al. (2011) A p53/miRNA-34 axis regulates Snail1-
dependent cancer cell epithelial-mesenchymal transition. J Cell Biol 195: 417–433. doi: 10.1083/jcb.
201103097 PMID: 22024162
65.
Knouf EC, Garg K, Arroyo JD, Correa Y, Sarkar D, et al. (2012) An integrative genomic approach iden-
tifies p73 and p63 as activators of miR-200 microRNA family transcription. Nucleic Acids Res 40:
499–510. doi: 10.1093/nar/gkr731 PMID: 21917857
66.
Antonini D, Russo MT, De Rosa L, Gorrese M, Del Vecchio L, et al. (2010) Transcriptional repression
of miR-34 family contributes to p63-mediated cell cycle progression in epidermal cells. J Invest Der-
matol 130: 1249–1257. doi: 10.1038/jid.2009.438 PMID: 20090763
67.
Agostini M, Tucci P, Steinert JR, Shalom-Feuerstein R, Rouleau M, et al. (2011) microRNA-34a
regulates neurite outgrowth, spinal morphology, and function. Proc Natl Acad Sci U S A 108:
21099–21104. doi: 10.1073/pnas.1112063108 PMID: 22160706
68.
Saini S, Majid S, Yamamura S, Tabatabai L, Suh SO, et al. (2011) Regulatory Role of mir-203 in Pros-
tate Cancer Progression and Metastasis. Clin Cancer Res 17: 5287–5298. doi: 10.1158/1078-0432.
CCR-10-2619 PMID: 21159887
69.
Blandino G, Dobbelstein M (2004) p73 and p63: why do we still need them? Cell Cycle 3: 886–894.
PMID: 15254416
70.
Dötsch V, Bernassola F, Coutandin D, Candi E, Melino G (2010) p63 and p73, the ancestors of p53.
Cold Spring Harb Perspect Biol 2: a004887. doi: 10.1101/cshperspect.a004887 PMID: 20484388
71.
Allocati N, Di Ilio C, De Laurenzi V (2012) p63/p73 in the control of cell cycle and cell death. Exp Cell
Res 318: 1285–1290. doi: 10.1016/j.yexcr.2012.01.023 PMID: 22326462
72.
Dotto GP (2009) Crosstalk of Notch with p53 and p63 in cancer growth control. Nat Rev Cancer 9:
587–595. doi: 10.1038/nrc2675 PMID: 19609265
73.
Moro L, Arbini AA, Yao JL, di Sant’Agnese PA, Marra E, et al. (2009) Mitochondrial DNA depletion in
prostate epithelial cells promotes anoikis resistance and invasion through activation of PI3K/Akt2.
Cell Death Differ 16: 571–583. doi: 10.1038/cdd.2008.178 PMID: 19079138
74.
Forster N, Ellisen LW (2011) Notch signaling mediates p63-induced quiescence: a new facet of p63/
Notch crosstalk. Cell Cycle 10: 3632–3633. doi: 10.4161/cc.10.21.18182 PMID: 22041817
75.
Zdzalik M, Pustelny K, Kedracka-Krok S, Huben K, Pecak A, et al. (2010) Interaction of regulators
Mdm2 and Mdmx with transcription factors p53, p63 and p73. Cell Cycle 9: 4584–4591. PMID:
21088494
76.
Feng Z, Levine AJ (2010) The regulation of energy metabolism and the IGF-1/mTOR pathways by the
p53 protein. Trends Cell Biol 20: 427–434. doi: 10.1016/j.tcb.2010.03.004 PMID: 20399660
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
25 / 29
77.
Kubo N, Okoshi R, Nakashima K, Shimozato O, Nakagawara A, et al. (2010) MDM2 promotes the pro-
teasomal degradation of p73 through the interaction with Itch in HeLa cells. Biochem Biophys Res
Commun 403: 405–411. doi: 10.1016/j.bbrc.2010.11.043 PMID: 21093410
78.
Zhang X, Tang N, Hadden TJ, Rishi AK (2011) Akt, FoxO and regulation of apoptosis. Biochim Bio-
phys Acta 1813: 1978–1986. doi: 10.1016/j.bbamcr.2011.03.010 PMID: 21440011
79.
Xu J, Zhou J-Y, Wei W-Z, Wu GS (2010) Activation of the Akt survival pathway contributes to TRAIL
resistance in cancer cells. PLoS One 5: e10226. doi: 10.1371/journal.pone.0010226 PMID:
20419107
80.
Li Y, Dowbenko D, Lasky LA (2002) AKT/PKB phosphorylation of p21Cip/WAF1 enhances protein
stability of p21Cip/WAF1 and promotes cell survival. J Biol Chem 277: 11352–11361. doi: 10.1074/
jbc.M109062200 PMID: 11756412
81.
Iliopoulos D, Polytarchou C, Hatziapostolou M, Kottakis F, Maroulakou IG, et al. (2009) MicroRNAs
differentially regulated by Akt isoforms control EMT and stem cell renewal in cancer cells. Sci Signal
2: ra62. doi: 10.1126/scisignal.2000356 PMID: 19825827
82.
Chin YR, Toker A (2010) The actin-bundling protein palladin is an Akt1-specific substrate that regu-
lates breast cancer cell migration. Mol Cell 38: 333–344. doi: 10.1016/j.molcel.2010.02.031 PMID:
20471940
83.
Chin YR, Toker A (2011) Akt isoform-specific signaling in breast cancer: uncovering an anti-migratory
role for palladin. Cell Adh Migr 5: 211–214. PMID: 21519185
84.
Liu H, Radisky DC, Nelson CM, Zhang H, Fata JE, et al. (2006) Mechanism of Akt1 inhibition of breast
cancer cell invasion reveals a protumorigenic role for TSC2. Proc Natl Acad Sci U S A 103: 4134–4139.
doi: 10.1073/pnas.0511342103 PMID: 16537497
85.
Héron-Milhavet L, Franckhauser C, Rana V, Berthenet C, Fisher D, et al. (2006) Only Akt1 is required
for proliferation, while Akt2 promotes cell cycle exit through p21 binding. Mol Cell Biol 26: 8267–8280.
doi: 10.1128/MCB.00201-06 PMID: 16982699
86.
Heron-Milhavet L, Franckhauser C, Fernandez A, Lamb NJ (2013) Characterization of the Akt2
domain essential for binding nuclear p21cip1 to promote cell cycle arrest during myogenic differentia-
tion. PLoS One 8: e76987. doi: 10.1371/journal.pone.0076987 PMID: 24194853
87.
Liao Y, Hung M-C (2010) Physiological regulation of Akt activity and stability. Am J Transl Res 2:
19–42. PMID: 20182580
88.
Talapatra S, Thompson CB (2001) Growth factor signaling in cell survival: implications for cancer
treatment. J Pharmacol Exp Ther 298: 873–878. PMID: 11504779
89.
Cabello-Verrugio C, Brandan E (2007) A novel modulatory mechanism of transforming growth factor-
beta signaling through decorin and LRP-1. J Biol Chem 282: 18842–18850. doi: 10.1074/jbc.
M700243200 PMID: 17485468
90.
Takeuchi Y, Kodama Y, Matsumoto T (1994) Bone matrix decorin binds transforming growth factor-
beta and enhances its bioactivity. J Biol Chem 269: 32634–32638. PMID: 7798269
91.
Okamoto O, Fujiwara S, Abe M, Sato Y (1999) Dermatopontin interacts with transforming growth fac-
tor beta and enhances its biological activity. Biochem J 337 (Pt 3: 537–541. PMID: 9895299
92.
Kopan R (2012) Notch signaling. Cold Spring Harb Perspect Biol 4. doi: 10.1101/cshperspect.
a011213
93.
Wu F, Stutzman A, Mo Y-Y (2007) Notch signaling and its role in breast cancer. Front Biosci 12:
4370–4383. PMID: 17485381
94.
Gonzalez AG, Naldi A, Sánchez L, Thieffry D, Chaouiya C (2006) GINsim: a software suite for the
qualitative modelling, simulation and analysis of regulatory networks. Biosystems 84: 91–100. doi:
10.1016/j.biosystems.2005.10.003 PMID: 16434137
95.
Wang H, Wu Q, Liu Z, Luo X, Fan Y, et al. (2014) Downregulation of FAP suppresses cell proliferation
and metastasis through PTEN/PI3K/AKT and Ras-ERK signaling in oral squamous cell carcinoma.
Cell Death Dis 5: e1155. doi: 10.1038/cddis.2014.122 PMID: 24722280
96.
Zhang L, Teng Y, Zhang Y, Liu J, Xu L, et al. (2012) C-Src-mediated RANKL-induced breast cancer
cell migration by activation of the ERK and Akt pathway. Oncol Lett 3: 395–400. doi: 10.3892/ol.2011.
487 PMID: 22740919
97.
Zhan Y, Kim S, Izumi Y, Izumiya Y, Nakao T, et al. (2003) Role of JNK, p38, and ERK in platelet-
derived growth factor-induced vascular proliferation, migration, and gene expression. Arterioscler
Thromb Vasc Biol 23: 795–801. doi: 10.1161/01.ATV.0000066132.32063.F2 PMID: 12637337
98.
To MD, Wong CE, Karnezis AN, Del Rosario R, Di Lauro R, et al. (2008) Kras regulatory elements
and exon 4A determine mutation specificity in lung cancer. Nat Genet 40: 1240–1244. doi: 10.1038/
ng.211 PMID: 18758463
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
26 / 29
99.
Zhang Z, Wang Y, Vikis HG, Johnson L, Liu G, et al. (2001) Wildtype Kras2 can inhibit lung carcino-
genesis in mice. Nat Genet 29: 25–33. doi: 10.1038/ng721 PMID: 11528387
100.
Li H, Cao H-F, Wan J, Li Y, Zhu M-L, et al. (2007) Growth inhibitory effect of wild-type Kras2 gene on a
colonic adenocarcinoma cell line. World J Gastroenterol 13: 934–938. PMID: 17352027
101.
Vartanian S, Bentley C, Brauer MJ, Li L, Shirasawa S, et al. (2013) Identification of mutant K-Ras-
dependent phenotypes using a panel of isogenic cell lines. J Biol Chem 288: 2403–2413. doi: 10.
1074/jbc.M112.394130 PMID: 23188824
102.
Benet M, Dulman RY, Suzme R, de Miera EV-S, Vega ME, et al. (2012) Wild type N-ras displays anti-
malignant properties, in part by downregulating decorin. J Cell Physiol 227: 2341–2351. doi: 10.1002/
jcp.22969 PMID: 21809347
103.
Xu J, Haigis KM, Firestone AJ, McNerney ME, Li Q, et al. (2013) Dominant role of oncogene dosage
and absence of tumor suppressor activity in Nras-driven hematopoietic transformation. Cancer Discov
3: 993–1001. doi: 10.1158/2159-8290.CD-13-0096 PMID: 23733505
104.
Molinari F, Frattini M (2013) Functions and Regulation of the PTEN Gene in Colorectal Cancer. Front
Oncol 3: 326. doi: 10.3389/fonc.2013.00326 PMID: 24475377
105.
Aguissa-Touré A-H, Li G (2012) Genetic alterations of PTEN in human melanoma. Cell Mol Life Sci
69: 1475–1491. doi: 10.1007/s00018-011-0878-0 PMID: 22076652
106.
McDowell KA, Riggins GJ, Gallia GL (2011) Targeting the AKT pathway in glioblastoma. Curr Pharm
Des 17: 2411–2420. PMID: 21827416
107.
Sun Z, Huang C, He J, Lamb KL, Kang X, et al. (2014) PTEN C-terminal deletion causes genomic
instability and tumor development. Cell Rep 6: 844–854. doi: 10.1016/j.celrep.2014.01.030 PMID:
24561254
108.
Mukherjee A, Karmakar P (2013) Attenuation of PTEN perturbs genomic stability via activation of Akt
and down-regulation of Rad51 in human embryonic kidney cells. Mol Carcinog 52: 611–618. doi: 10.
1002/mc.21903 PMID: 22488521
109.
He L, Hou X, Kanel G, Zeng N, Galicia V, et al. (2010) The critical role of AKT2 in hepatic steatosis
induced by PTEN loss. Am J Pathol 176: 2302–2308. doi: 10.2353/ajpath.2010.090931 PMID:
20348245
110.
Al-Saad S, Donnem T, Al-Shibli K, Persson M, Bremnes RM, et al. (2009) Diverse prognostic roles of
Akt isoforms, PTEN and PI3K in tumor epithelial cells and stromal compartment in non-small cell lung
cancer. Anticancer Res 29: 4175–4183. PMID: 19846969
111.
Trotman LC, Alimonti A, Scaglioni PP, Koutcher JA, Cordon-Cardo C, et al. (2006) Identification of a
tumour suppressor network opposing nuclear Akt function. Nature 441: 523–527. doi: 10.1038/
nature04809 PMID: 16680151
112.
Chin YMR, Yuan X, Balk SP, Toker A (2014) Pten-deficient tumors depend on akt2 for maintenance
and survival. Cancer Discov. doi: 10.1158/2159-8290.CD-13-0873
113.
Wickremasinghe RG, Prentice AG, Steele AJ (2011) p53 and Notch signaling in chronic lymphocytic
leukemia: clues to identifying novel therapeutic strategies. Leuk Off J Leuk Soc Am Leuk Res Fund,
UK 25: 1400–1407. doi: 10.1038/leu.2011.103
114.
Hoshino Y, Katsuno Y, Ehata S, Miyazono K (2011) Autocrine TGF-β protects breast cancer cells
from apoptosis through reduction of BH3-only protein, Bim. J Biochem 149: 55–65. doi: 10.1093/jb/
mvq114 PMID: 20880961
115.
Xiao M, Liu YG, Zou MC, Zou F (2014) Sodium butyrate induces apoptosis of human colon cancer
cells by modulating ERK and sphingosine kinase 2. Biomed Environ Sci 27: 197–203. doi: 10.3967/
bes2014.040 PMID: 24709100
116.
Sayan AE, Griffiths TR, Pal R, Browne GJ, Ruddick A, et al. (2009) SIP1 protein protects cells from
DNA damage-induced apoptosis and has independent prognostic value in bladder cancer. Proc Natl
Acad Sci U S A 106: 14884–14889. doi: 10.1073/pnas.0902042106 PMID: 19706487
117.
Sahlberg SH, Gustafsson A-S, Pendekanti PN, Glimelius B, Stenerlöw B (2014) The influence of AKT
isoforms on radiation sensitivity and DNA repair in colon cancer cell lines. Tumour Biol 35: 3525–3534.
doi: 10.1007/s13277-013-1465-9 PMID: 24338765
118.
Daroqui MC, Vazquez P, Bal de Kier Joffé E, Bakin A V, Puricelli LI (2012) TGF-β autocrine pathway
and MAPK signaling promote cell invasiveness and in vivo mammary adenocarcinoma tumor progres-
sion. Oncol Rep 28: 567–575. doi: 10.3892/or.2012.1813 PMID: 22614218
119.
Gomes LR, Terra LF, Wailemann RA, Labriola L, Sogayar MC (2012) TGF-β1 modulates the homeo-
stasis between MMPs and MMP inhibitors through p38 MAPK and ERK1/2 in highly invasive breast
cancer cells. BMC Cancer 12: 26. doi: 10.1186/1471-2407-12-26 PMID: 22260435
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
27 / 29
120.
Tan TZ, Miow QH, Miki Y, Noda T, Mori S, et al. (2014) Epithelial-mesenchymal transition spectrum
quantification and its efficacy in deciphering survival and drug responses of cancer patients. EMBO
Mol Med 6: 1279–1293. doi: 10.15252/emmm.201404208 PMID: 25214461
121.
Zhou H-M, Dong T-T, Wang L-L, Feng B, Zhao H-C, et al. (2012) Suppression of colorectal cancer
metastasis by nigericin through inhibition of epithelial-mesenchymal transition. World J Gastroenterol
18: 2640–2648. doi: 10.3748/wjg.v18.i21.2640 PMID: 22690072
122.
Wendt MK, Taylor MA, Schiemann BJ, Schiemann WP (2011) Down-regulation of epithelial cadherin
is required to initiate metastatic outgrowth of breast cancer. Mol Biol Cell 22: 2423–2435. doi: 10.
1091/mbc.E11-04-0306 PMID: 21613543
123.
Stoll G, Viara E, Barillot E, Calzone L (2012) Continuous time Boolean modeling for biological signal-
ing: application of Gillespie algorithm. BMC Syst Biol 6: 116. doi: 10.1186/1752-0509-6-116 PMID:
22932419
124.
Muller PAJ, Vousden KH, Norman JC (2011) p53 and its mutants in tumor cell migration and invasion.
J Cell Biol 192: 209–218. doi: 10.1083/jcb.201009059 PMID: 21263025
125.
Wang XQ, Ongkeko WM, Lau AW, Leung KM, Poon RY (2001) A possible role of p73 on the modula-
tion of p53 level through MDM2. Cancer Res 61: 1598–1603. PMID: 11245471
126.
Wang Z, Li Y, Kong D, Banerjee S, Ahmad A, et al. (2009) Acquisition of epithelial-mesenchymal tran-
sition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch
signaling pathway. Cancer Res 69: 2400–2407. doi: 10.1158/0008-5472.CAN-08-4312 PMID:
19276344
127.
Poon IKH, Lucas CD, Rossi AG, Ravichandran KS (2014) Apoptotic cell clearance: basic biology and
therapeutic potential. Nat Rev Immunol 14: 166–180. doi: 10.1038/nri3607 PMID: 24481336
128.
Paterson EL, Kazenwadel J, Bert AG, Khew-Goodall Y, Ruszkiewicz A, et al. (2013) Down-regulation
of the miRNA-200 family at the invasive front of colorectal cancers with degraded basement mem-
brane indicates EMT is involved in cancer progression. Neoplasia 15: 180–191. PMID: 23441132
129.
El Marjou F, Janssen K-P, Chang BH-J, Li M, Hindie V, et al. (2004) Tissue-specific and inducible
Cre-mediated recombination in the gut epithelium. Genesis 39: 186–193. doi: 10.1002/gene.20042
PMID: 15282745
130.
Murtaugh LC, Stanger BZ, Kwan KM, Melton DA (2003) Notch signaling controls multiple steps of
pancreatic differentiation. Proc Natl Acad Sci U S A 100: 14920–14925. doi: 10.1073/pnas.
2436557100 PMID: 14657333
131.
Herbst A, Jurinovic V, Krebs S, Thieme SE, Blum H, et al. (2014) Comprehensive analysis of β-
catenin target genes in colorectal carcinoma cell lines with deregulated Wnt/β-catenin signaling. BMC
Genomics 15: 74. doi: 10.1186/1471-2164-15-74 PMID: 24467841
132.
Zeilstra J, Joosten SPJ, Wensveen FM, Dessing MC, Schütze DM, et al. (2011) WNT signaling con-
trols expression of pro-apoptotic BOK and BAX in intestinal cancer. Biochem Biophys Res Commun
406: 1–6. doi: 10.1016/j.bbrc.2010.12.070 PMID: 21184732
133.
Prosperi JR, Goss KH (2010) A Wnt-ow of opportunity: targeting the Wnt/beta-catenin pathway in
breast cancer. Curr Drug Targets 11: 1074–1088. PMID: 20545611
134.
Buda A, Pignatelli M (2011) E-cadherin and the cytoskeletal network in colorectal cancer development
and metastasis. Cell Commun Adhes 18: 133–143. doi: 10.3109/15419061.2011.636465 PMID:
22176698
135.
Wu K, Fan J, Zhang L, Ning Z, Zeng J, et al. (2012) PI3K/Akt to GSK3β/β-catenin signaling cascade
coordinates cell colonization for bladder cancer bone metastasis through regulating ZEB1 transcrip-
tion. Cell Signal 24: 2273–2282. doi: 10.1016/j.cellsig.2012.08.004 PMID: 22906492
136.
Morán A, Ortega P, de Juan C, Fernández-Marcelo T, Frías C, et al. (2010) Differential colorectal car-
cinogenesis: Molecular basis and clinical relevance. World J Gastrointest Oncol 2: 151–158. doi: 10.
4251/wjgo.v2.i3.151 PMID: 21160823
137.
Yuzugullu H, Benhaj K, Ozturk N, Senturk S, Celik E, et al. (2009) Canonical Wnt signaling is antago-
nized by noncanonical Wnt5a in hepatocellular carcinoma cells. Mol Cancer 8: 90. doi: 10.1186/
1476-4598-8-90 PMID: 19849855
138.
Pang H, Lu H, Song H, Meng Q, Zhao Y, et al. (2013) Prognostic values of osteopontin-c, E-cadherin
and β-catenin in breast cancer. Cancer Epidemiol 37: 985–992. doi: 10.1016/j.canep.2013.08.005
PMID: 24012693
139.
Zhang H, Liu J, Yue D, Gao L, Wang D, et al. (2013) Clinical significance of E-cadherin, β-catenin,
vimentin and S100A4 expression in completely resected squamous cell lung carcinoma. J Clin Pathol
66: 937–945. doi: 10.1136/jclinpath-2013-201467 PMID: 23853314
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
28 / 29
140.
Chiu CG, Chan SK, Fang ZA, Masoudi H, Wood-Baker R, et al. (2012) Beta-catenin expression is
prognostic of improved non-small cell lung cancer survival. Am J Surg 203: 654–659. doi: 10.1016/j.
amjsurg.2012.01.002 PMID: 22402266
141.
Nozawa N, Hashimoto S, Nakashima Y, Matsuo Y, Koga T, et al. (2006) Immunohistochemical alpha-
and beta-catenin and E-cadherin expression and their clinicopathological significance in human lung
adenocarcinoma. Pathol Res Pract 202: 639–650. doi: 10.1016/j.prp.2006.03.007 PMID: 16843618
142.
Morikawa T, Kuchiba A, Yamauchi M, Meyerhardt JA, Shima K, et al. (2011) Association of CTNNB1
(beta-catenin) alterations, body mass index, and physical activity with survival in patients with colorec-
tal cancer. JAMA 305: 1685–1694. doi: 10.1001/jama.2011.513 PMID: 21521850
143.
Morikawa T, Kuchiba A, Lochhead P, Nishihara R, Yamauchi M, et al. (2013) Prospective analysis of
body mass index, physical activity, and colorectal cancer risk associated with β-catenin (CTNNB1)
status. Cancer Res 73: 1600–1610. doi: 10.1158/0008-5472.CAN-12-2276 PMID: 23442321
144.
Bruun J, Kolberg M, Nesland JM, Svindland A, Nesbakken A, et al. (2014) Prognostic Significance of
β-Catenin, E-Cadherin, and SOX9 in Colorectal Cancer: Results from a Large Population-Represen-
tative Series. Front Oncol 4: 118. doi: 10.3389/fonc.2014.00118 PMID: 24904831
145.
Pourreyron C, Reilly L, Proby C, Panteleyev A, Fleming C, et al. (2012) Wnt5a is strongly expressed
at the leading edge in non-melanoma skin cancer, forming active gradients, while canonical Wnt sig-
nalling is repressed. PLoS One 7: e31827. doi: 10.1371/journal.pone.0031827 PMID: 22384081
146.
Mikels AJ, Nusse R (2006) Purified Wnt5a protein activates or inhibits beta-catenin-TCF signaling
depending on receptor context. PLoS Biol 4: e115. doi: 10.1371/journal.pbio.0040115 PMID:
16602827
147.
Yuan Y, Niu CC, Deng G, Li ZQ, Pan J, et al. (2011) The Wnt5a/Ror2 noncanonical signaling pathway
inhibits canonical Wnt signaling in K562 cells. Int J Mol Med 27: 63–69. doi: 10.3892/ijmm.2010.560
PMID: 21069266
148.
Wang L, Liu X, Gusev E, Wang C, Fagotto F (2014) Regulation of the phosphorylation and nuclear
import and export of β-catenin by APC and its cancer-related truncated form. J Cell Sci 127:
1647–1659. doi: 10.1242/jcs.131045 PMID: 24496450
149.
Roy HK, Olusola BF, Clemens DL, Karolski WJ, Ratashak A, et al. (2002) AKT proto-oncogene over-
expression is an early event during sporadic colon carcinogenesis. Carcinogenesis 23: 201–205.
PMID: 11756242
150.
Rychahou PG, Kang J, Gulhati P, Doan HQ, Chen LA, et al. (2008) Akt2 overexpression plays a criti-
cal role in the establishment of colorectal cancer metastasis. Proc Natl Acad Sci U S A 105:
20315–20320. doi: 10.1073/pnas.0810715105 PMID: 19075230
151.
Steinway SN, Gomez Tejeda Zañudo J, Ding W, Rountree CB, Feith DJ, et al. (2014) Network model-
ing of TGFβ signaling in hepatocellular carcinoma epithelial-to-mesenchymal transition reveals joint
Sonic hedgehog and Wnt pathway activation. Cancer Res. doi: 10.1158/0008-5472.CAN-14-0225
152.
Nakabayashi H, Taketa K, Miyano K, Yamane T, Sato J (1982) Growth of human hepatoma cells lines
with differentiated functions in chemically defined medium. Cancer Res 42: 3858–3863. PMID:
6286115
153.
Alexander JJ, Bey EM, Geddes EW, Lecatsas G (1976) Establishment of a continuously growing cell
line from primary carcinoma of the liver. S Afr Med J 50: 2124–2128. PMID: 63998
154.
Lin Y, Shi CY, Li B, Soo BH, Mohammed-Ali S, et al. (1996) Tumour suppressor p53 and Rb genes in
human hepatocellular carcinoma. Ann Acad Med Singapore 25: 22–30. PMID: 8779541
155.
Austinat M, Dunsch R, Wittekind C, Tannapfel A, Gebhardt R, et al. (2008) Correlation between beta-
catenin mutations and expression of Wnt-signaling target genes in hepatocellular carcinoma. Mol
Cancer 7: 21. doi: 10.1186/1476-4598-7-21 PMID: 18282277
Modelling of Metastasis Process
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004571
November 3, 2015
29 / 29
|
26528548
|
EMT = ( ( CDH2 ) AND NOT ( CDH1 ) )
p21 = ( ( ( SMAD AND ( ( ( NICD ) ) ) ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) ) OR ( ( ( p63 ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) ) OR ( ( ( p73 ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) ) OR ( ( ( AKT2 ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) ) OR ( ( ( p53 ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) )
p53 = ( ( ( ( ( NICD ) AND NOT ( p73 ) ) AND NOT ( AKT2 ) ) AND NOT ( SNAI2 ) ) AND NOT ( AKT1 ) ) OR ( ( ( ( ( CTNNB1 ) AND NOT ( p73 ) ) AND NOT ( AKT2 ) ) AND NOT ( SNAI2 ) ) AND NOT ( AKT1 ) ) OR ( ( ( ( ( DNAdamage ) AND NOT ( p73 ) ) AND NOT ( AKT2 ) ) AND NOT ( SNAI2 ) ) AND NOT ( AKT1 ) ) OR ( ( ( ( ( miR34 ) AND NOT ( p73 ) ) AND NOT ( AKT2 ) ) AND NOT ( SNAI2 ) ) AND NOT ( AKT1 ) )
p63 = ( ( ( ( ( ( DNAdamage ) AND NOT ( AKT2 ) ) AND NOT ( NICD ) ) AND NOT ( AKT1 ) ) AND NOT ( p53 ) ) AND NOT ( miR203 ) )
NICD = ( ( ( ( ( ( ECM ) AND NOT ( p73 ) ) AND NOT ( miR200 ) ) AND NOT ( miR34 ) ) AND NOT ( p53 ) ) AND NOT ( p63 ) )
Invasion = ( SMAD AND ( ( ( CDH2 ) ) ) ) OR ( CTNNB1 )
AKT2 = ( TWIST1 AND ( ( ( TGFbeta OR CDH2 OR GF ) AND ( ( ( NOT miR34 AND NOT miR203 AND NOT p53 ) ) ) ) ) )
AKT1 = ( ( ( ( CTNNB1 AND ( ( ( TGFbeta ) ) OR ( ( CDH2 ) ) OR ( ( NICD ) ) OR ( ( GF ) ) ) ) AND NOT ( CDH1 ) ) AND NOT ( p53 ) ) AND NOT ( miR34 ) )
miR200 = ( ( ( ( ( ( p63 ) AND NOT ( SNAI2 ) ) AND NOT ( AKT2 ) ) AND NOT ( SNAI1 ) ) AND NOT ( ZEB2 ) ) AND NOT ( ZEB1 ) ) OR ( ( ( ( ( ( p73 ) AND NOT ( SNAI2 ) ) AND NOT ( AKT2 ) ) AND NOT ( SNAI1 ) ) AND NOT ( ZEB2 ) ) AND NOT ( ZEB1 ) ) OR ( ( ( ( ( ( p53 ) AND NOT ( SNAI2 ) ) AND NOT ( AKT2 ) ) AND NOT ( SNAI1 ) ) AND NOT ( ZEB2 ) ) AND NOT ( ZEB1 ) )
miR34 = ( ( ( AKT2 AND ( ( ( NOT ZEB2 AND NOT SNAI1 AND NOT ZEB1 ) AND ( ( ( p73 OR p53 ) ) ) ) ) ) AND NOT ( p63 ) ) AND NOT ( AKT1 ) )
Metastasis = ( Migration )
SNAI1 = ( ( ( ( ( NICD ) AND NOT ( miR203 ) ) AND NOT ( CTNNB1 ) ) AND NOT ( miR34 ) ) AND NOT ( p53 ) ) OR ( ( ( ( ( TWIST1 ) AND NOT ( miR203 ) ) AND NOT ( CTNNB1 ) ) AND NOT ( miR34 ) ) AND NOT ( p53 ) )
VIM = ( ZEB2 ) OR ( CTNNB1 )
ZEB2 = ( ( ( NICD ) AND NOT ( miR203 ) ) AND NOT ( miR200 ) ) OR ( ( ( SNAI1 ) AND NOT ( miR203 ) ) AND NOT ( miR200 ) ) OR ( ( ( SNAI2 AND ( ( ( TWIST1 ) ) ) ) AND NOT ( miR203 ) ) AND NOT ( miR200 ) )
ZEB1 = ( ( NICD ) AND NOT ( miR200 ) ) OR ( ( SNAI2 ) AND NOT ( miR200 ) ) OR ( ( TWIST1 AND ( ( ( SNAI1 ) ) ) ) AND NOT ( miR200 ) ) OR ( ( CTNNB1 ) AND NOT ( miR200 ) )
TGFbeta = ( ( ECM ) AND NOT ( CTNNB1 ) ) OR ( ( NICD ) AND NOT ( CTNNB1 ) )
CDH2 = ( TWIST1 )
DKK1 = ( NICD ) OR ( CTNNB1 )
TWIST1 = ( SNAI1 ) OR ( NICD ) OR ( CTNNB1 )
ERK = ( ( NICD ) AND NOT ( AKT1 ) ) OR ( ( CDH2 ) AND NOT ( AKT1 ) ) OR ( ( GF ) AND NOT ( AKT1 ) ) OR ( ( SMAD ) AND NOT ( AKT1 ) )
SMAD = ( ( ( TGFbeta ) AND NOT ( miR203 ) ) AND NOT ( miR200 ) )
CellCycleArrest = ( ( miR200 ) AND NOT ( AKT1 ) ) OR ( ( miR203 ) AND NOT ( AKT1 ) ) OR ( ( ZEB2 ) AND NOT ( AKT1 ) ) OR ( ( p21 ) AND NOT ( AKT1 ) ) OR ( ( miR34 ) AND NOT ( AKT1 ) )
miR203 = ( ( ( ( p53 ) AND NOT ( ZEB1 ) ) AND NOT ( SNAI1 ) ) AND NOT ( ZEB2 ) )
Apoptosis = ( ( ( ( miR200 ) AND NOT ( ZEB2 ) ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) ) OR ( ( ( ( p63 ) AND NOT ( ZEB2 ) ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) ) OR ( ( ( ( p73 ) AND NOT ( ZEB2 ) ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) ) OR ( ( ( ( miR34 ) AND NOT ( ZEB2 ) ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) ) OR ( ( ( ( p53 ) AND NOT ( ZEB2 ) ) AND NOT ( ERK ) ) AND NOT ( AKT1 ) )
SNAI2 = ( ( ( ( NICD ) AND NOT ( miR200 ) ) AND NOT ( miR203 ) ) AND NOT ( p53 ) ) OR ( ( ( ( CTNNB1 ) AND NOT ( miR200 ) ) AND NOT ( miR203 ) ) AND NOT ( p53 ) ) OR ( ( ( ( TWIST1 ) AND NOT ( miR200 ) ) AND NOT ( miR203 ) ) AND NOT ( p53 ) )
GF = ( ( CDH2 ) AND NOT ( CDH1 ) ) OR ( ( GF ) AND NOT ( CDH1 ) )
CDH1 = NOT ( ( SNAI1 ) OR ( SNAI2 ) OR ( TWIST1 ) OR ( ZEB2 ) OR ( ZEB1 ) OR ( AKT2 ) )
Migration = ( ( ( ( VIM AND ( ( ( ERK AND AKT2 AND Invasion AND EMT ) ) ) ) AND NOT ( miR200 ) ) AND NOT ( AKT1 ) ) AND NOT ( p63 ) )
p73 = ( ( ( ( ( DNAdamage ) AND NOT ( p53 ) ) AND NOT ( AKT2 ) ) AND NOT ( AKT1 ) ) AND NOT ( ZEB1 ) )
CTNNB1 = NOT ( ( AKT1 ) OR ( miR200 ) OR ( p63 ) OR ( CDH2 ) OR ( CDH1 ) OR ( DKK1 ) OR ( p53 ) OR ( miR34 ) )
|
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
DOI 10.1186/s12976-015-0023-0
RESEARCH
Open Access
A Boolean network model of human
gonadal sex determination
Osiris Ríos1,2, Sara Frias1,3, Alfredo Rodríguez1,4, Susana Kofman5, Horacio Merchant3, Leda Torres1*
and Luis Mendoza3,6*
*Correspondence:
ledactorres@gmail.com;
lmendoza@biomedicas.unam.mx
1Instituto Nacional de Pediatría,
Laboratorio de Citogenética, Av.
Insurgentes Sur 3700 C, 04530
México City, México
3Instituto de Investigaciones
Biomédicas, UNAM, 04510 Mexico
City, México
Full list of author information is
available at the end of the article
Abstract
Background: Gonadal sex determination (GSD) in humans is a complex biological
process that takes place in early stages of embryonic development when the
bipotential gonadal primordium (BGP) differentiates towards testes or ovaries. This
decision is directed by one of two distinct pathways embedded in a GSD network
activated in a population of coelomic epithelial cells, the Sertoli progenitor cells (SPC)
and the granulosa progenitor cells (GPC). In males, the pathway is activated when the
Sex-Determining Region Y (SRY) gene starts to be expressed, whereas in females the
WNT4/β-catenin pathway promotes the differentiation of the GPCs towards ovaries.
The interactions and dynamics of the elements that constitute the GSD network are
poorly understood, thus our group is interested in inferring the general architecture of
this network as well as modeling the dynamic behavior of a set of genes associated to
this process under wild-type and mutant conditions.
Methods: We reconstructed the regulatory network of GSD with a set of genes
directly associated with the process of differentiation from SPC and GPC towards Sertoli
and granulosa cells, respectively. These genes are experimentally well-characterized
and the effects of their deficiency have been clinically reported. We modeled this GSD
network as a synchronous Boolean network model (BNM) and characterized its
attractors under wild-type and mutant conditions.
Results: Three attractors with a clear biological meaning were found; one of them
corresponding to the currently known gene expression pattern of Sertoli cells, the
second correlating to the granulosa cells and, the third resembling a disgenetic gonad.
Conclusions: The BNM of GSD that we present summarizes the experimental data on
the pathways for Sertoli and granulosa establishment and sheds light on the overall
behavior of a population of cells that differentiate within the developing gonad. With
this model we propose a set of regulatory interactions needed to activate either the SRY
or the WNT4/β-catenin pathway as well as their downstream targets, which are critical
for further sex differentiation. In addition, we observed a pattern of altered regulatory
interactions and their dynamics that lead to some disorders of sex development (DSD).
Keywords: Sex determination, Gonadal sex determination, Boolean model, Gene
regulatory network
© 2015 Ríos et al. Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International
License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any
medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons
license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.
org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 2 of 18
Background
Sex development is a complex biological process that occurs during the embryonic and
fetal stages of an individual. For a better understanding sex development is divided
into three consecutive steps: 1) chromosomal sex determination (CSD); 2) gonadal
sex determination (GSD); and 3) phenotypic sex differentiation (PSD). CSD is estab-
lished at conception when the complement of sex chromosomes, XX or XY, is received.
GSD, which is the process that we analyze in this study, refers to the set of genes
and their regulatory interactions that trigger the development toward testes or ovaries,
underlined by a gene regulatory network [1–4]. Finally, PSD involves the development
of the female and male internal and external genitalia in response to the hormones
secreted by the ovaries and testes. Both male and female PSD occur in two tempo-
ral phases, the first occurs within the fetus after GSD and the second occurs during
puberty [5, 6].
GSD occurs within a heterogeneously composed structure called bipotential gonadal
primordium (BGP). This structure, located on the ventromedial surface of the
mesonephros [5–7], is critical for sex development since it can differentiate either as
testes or ovaries [8]. The BGP originates the actual gonad that is composed by a) the
germinal cells (GCs), b) the steroidogenic somatic cells, such as the theca cells in ovary
and the Leydig cells in testis that produce stradiol and testosterone, respectively; and
c) the support somatic cells, including granulosa cells in ovary and Sertoli cells in
testis.
Sertoli and granulosa cells originate from a common population of coelomic epithelial
cells corresponding to the Sertoli progenitor cells (SPC) or granulosa progenitor cells
(GPC) that migrate towards the BGP [9, 10]. In males, the SPCs start to differentiate
toward Sertoli cells after 44 days of development (Carnegie-Stage 18). The mechanism
involves activation of the expression of the Sex-determining Region Y gene (SRY) that
codifies the SRY transcription factor [9, 11]. SRY associates with other transcription fac-
tors (i.e., CBX2, SF1) to regulate expression of the SOX9 gene that positively regulates the
expression of genes associated to Sertoli cells (i.e., SOX9, FGF9, PGD2, DHH, AMH) [2].
In females, where SRY is absent, GSD initiates after 49 days of development (Carnegie-
Stage 20). In this case, the GPCs of coelomic origin differentiate towards granulosa cells
by the action of a distinct gene regulatory pathway. Most likely, an increased amount of
the transcription factor β-catenin up-regulates a set of downstream genes associated to
granulosa, such as FOXL2 and RSPO1 [2, 12, 13]. Thus, the mechanism underlying GSD
involves a common population of undifferentiated cells with the potential to diverge into
two cell fates. The male pathway leads towards Sertoli cell fate determination and dif-
ferentiation, whereas the female pathway leads to granulosa cell fate determination and
differentiation.
Once differentiated, the Sertoli cells act as organizing centers, enclosing GCs to form
testicular cords and secreting factors such as DHH and PDGF, which are essential for
development of the fetal population of Leydig cells [14]. Granulosa cells are the female
equivalent of the Sertoli cells, as they enclose GCs and secrete factors necessary for
oocyte growth and maturation. The regulatory network controlling GSD and differentia-
tion toward Sertoli or granulosa cell consists, in a broad sense, of multiple target genes,
different types of RNAs, transcription factors, nuclear receptors and signaling molecules.
These elements are present in undifferentiated cells and interact in a concerted way either
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 3 of 18
activating or repressing target genes at the time of GSD to balance the fate toward Sertoli
or granulosa cells [15–17].
The total number of genes implicated in the regulatory network of GSD of humans
and mammals remains elusive, as well as their complete regulatory interactions and their
effects on the process of Sertoli or granulosa cells differentiation. However, it is well
known that mutations in their components underlay the so-called disorders of sex devel-
opment (DSD), a series of genetic conditions characterized by anomalies in gonads as
well as in internal and external genitalia. The incidence of DSDs, as estimated by the
The Lawson Wilkins Pediatric Endocrine Society (LWPES) and the European Society for
Pediatric Endocrinology (ESPE), is 1 in 4,500 births [18] and can be attributed to muta-
tions in various genes of the GSD network. For example, mutations in CBX2, GATA4 and
WT1 genes result in a wide range of phenotypic alterations characterized by ambiguous
or female external genitalia with the presence or absence of Mullerian structures in 46,XY
DSDs patients [19]. In contrast, 46,XX DSDs cause masculinization of the female fetus
(normal males with no ovarian tissue) [20]. In other cases 46,XX DSD patients have a
female phenotype but fail to develop ovaries, presenting instead a “streak gonad”? (streaks
of connective fibrous tissue) [21].
Boolean network models (BNM) are formal tools for analyzing the structure and
dynamic behavior of genetic regulatory networks. BNM are best suited for describing
poorly-characterized systems with no or few kinetic details, such as the GSD network.
These models represent molecular entities (genes, transcription factors and RNAs) as
nodes interacting among them within a network. Each node can have only two qualitative
states: 0 (OFF) and 1 (ON). The OFF state is equivalent to a below-threshold concen-
tration or activity, which is insufficient to initiate the intended process or regulation,
while the ON state is equivalent to an above-threshold concentration or activity [22]. The
ON/OFF state of a node within the network is determined by a Boolean function that
encompasses the known regulatory elements of the target node (transcription factors,
nuclear receptors, signaling molecules). The state of these regulatory elements is updated
over consecutive time steps of a simulation until the system converges to either a steady
state or a cycle. BNMs describe the dynamic state of the nodes in a network by updating
the state of the nodes according with the set of regulatory functions [23]. BNMs have been
implemented for the analysis of developmental programs such as flower morphogenesis
in A. thaliana [24], early cardiac development in mice [25], and expression pattern of the
segment polarity genes in Drosophila [26] to name a few.
Despite the relatively high incidence of DSDs, their molecular basis at the level of the
regulatory network remain poorly understood. Thus, we are interested in constructing
a BNM of the process of gonadal sex determination with an emphasis on the regulatory
elements that are present at early stages of development and control the differentiation of
SCP and GCP towards Sertoli and granulosa cells, respectively, allowing us to analyze the
origin of some DSDs. For in-depth reviews about the genes involved in GSD and DSDs
see: [18, 19, 27, 28], as well as the list of genes and interactions in the Additional file 1 of
the supplementary information.
In this study we present a BNM that describes the dynamics of the GSD regulatory
network starting from the UGR until Sertoli/granulosa cells differentiation. The pro-
posed regulatory network incorporates a large amount of published information related
to functional interactions among the genes involved in this process, while the BNM of
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 4 of 18
GSD describes the dynamics of the elements contained within the network under wild-
type and mutant conditions. With the current model we explore a formal description of
the functional relationships among the genes and gene products associated to GSD, and
generate some predictions about the expected regulatory behavior under wild-type or
altered conditions within elements of the UGR and elements of the bipotential gonadal
primordium such as CBX2, GATA4, and WT1. Additional predictions are indicated in
the female pathway where the transcription factor β-catenin seems to play an impor-
tant role in the activation of female-specific genes (for example, WNT4, RSPO1, and
FOXL2).
Methods
The network of gonadal sex determination
To construct the network we selected a set of genes with well-known clinical and
experimental data demonstrating their association to GSD under wild-type and mutant
conditions. The genes, depicted in a regulatory diagram (Fig. 1), include: CBX2, NR5A1,
GATA4, WT1pKTS, WT1mKTS, NR0B1, SRY, SOX9, FGF9, PGD2, DHH, AMH, DKK1,
DMRT1, CTNNB1, WNT4, FOXL2, RSPO1, and a special node called UGR. The inter-
actions among these nodes are denoted by edges. We distinguish positive interactions
(activation) by connecting two nodes with an arrow-head line. Negative interactions
(inhibition) are denoted by connecting two nodes with a bar-head line.
The regulatory interactions among nodes were inferred, with emphasis in humans,
from: (1) clinical studies of patients with DSDs, which carried mutations on sex deter-
mining genes; (2) genetic expression patterns associated to GSD (between 5th and 8th
weeks of embryonic development); and (3) molecular evidence of interactions at the
level of transcriptional regulation of target genes (i.e., up or down regulation of a tar-
get gene by means of protein-DNA interactions under wild type, mutated or transgenic
constructs). Experimental evidence on mice was integrated into the network when neces-
sary, especially in the female pathway where human information is lacking. References to
clinical and experimental data can be found in the Additional file 1 of the Supplementary
Information.
The network includes the special UGR node, representing the urogenital ridge, an
embryonic structure precursor of the nephrogenic cord and gonads. The UGR node
encompasses the following genes: LHX1, LHX9, EMX2, PAX2 and, PAX8. Although
expression of these genes is essential for growth and maintenance of the UGR, little
evidence was found in human, as well as in mouse, about their specific regulatory inter-
actions, thus these genes were grouped within the UGR node, since mutations in any of
these genes impair subsequent gonadal development [29–31].
The pathway towards pre-Sertoli or pre-granulosa cells is shown in Fig. 1. The blue
nodes correspond to Sertoli cell fate determination pathway and include: SRY, SOX9,
FGF9, PGD2, DHH, AMH, DKK1 and DMRT1 nodes, whereas the pink nodes correspond
to granulosa cell fate determination pathway including: CTNNB1, WNT4, FOXL2 and
RSPO1. Notice that the granulosa cell fate determination was complemented with mice
information. For example, we considered the canonical Wnt4/β-catenin pathway as a key
regulatory element of female nodes within the network since relative expression of Fst,
Gng13, Foxl2, Irx3 and, Sp5 has been shown to be down-regulated when β-catenin is lost
in female mice in early stages of ovarian development [32].
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 5 of 18
Fig. 1 Network of Gonadal Sex Determination leading to Sertoli or granulosa cell fate commitment and
differentiation. The network was inferred from reviewed experimental evidence of genes associated within
the process and structured according to developmental stages in: urogenital ridge (UGR node), bipotential
gonadal primordium (yellow nodes), male pathway of sex determination (blue nodes), female pathway of sex
determination (pink nodes). Nodes represent genes; arrow lines denote activation; bar head lines indicate
inhibition; black, blue and, pink solid lines represent validated interactions in human; green solid lines
represent interactions validated in mouse; punctuated lines in orange represent model predictions
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 6 of 18
The network as a Boolean model
The process of GSD is poorly characterized at the quantitative level, i.e., kinetic informa-
tion regarding the interactions of the elements of this regulatory network is still lacking,
therefore the implementation of the GSD network as a continuous model is, at this
moment, out of reach. Given this, we decided to model the network as a discrete dynami-
cal system so as to describe the qualitative observations that are experimentally reported.
Specifically, we used a Boolean approach where every node might have one of two pos-
sible states; 1 (ON) or 0 (OFF), indicating that a given node within the network model is
active or inactive, respectively.
To determine the activation state of each node in the GSD model we translated the
experimental regulatory interactions into a set of Boolean functions with the use of the
logical operators AND, OR and NOT (Table 1). The logical operator AND is used if two
nodes named A and B are required to activate a third node named C. The logical operator
OR is used if two nodes named A or B can activate, by its own, node C. The logical oper-
ator NOT is used if node A is an inhibitor of node B. Thus, the state of a given node over
time is determined by the activation state of its regulators. We integrated to the model
additional regulatory interactions not reported by observational or experimental studies
(Table 2). These interactions were inferred from analysis of the dynamics of the Boolean
model and might be considered as model predictions that deserve further experimen-
tation to be validated. Interactions of model predictions are shown in Fig. 1 as orange
dashed lines.
We performed an initial exhaustive evaluation of the dynamic behavior of the wild
type model, simulating all possible initial activation states. Three fixed-point attractors
were obtained, and we performed a search focused in finding the state transitions cor-
responding to both male and female pathways. To recover the wild type “male pathway”,
we initiated the simulations with the UGR node in ON. In contrast, to created a wild
type “female pathway”, without the SRY node, we set the UGR and WNT4 nodes as active
Table 1 Set of functions for the Boolean model of gonadal sex determination
UGR, UGR & ! (NR5A1 | WNT4)
CBX2, UGR & ! (NR0B1 & WNT4 & CTNNB1)
GATA4, (UGR | WNT4 | NR5A1 | SRY)
WT1mKTS, (UGR | GATA4)
WT1pKTS, (UGR | GATA4) & ! (WNT4 & CTNNB1)
NR5A1, (UGR | CBX2 | WT1mKTS | GATA4) & ! (NR0B1 & WNT4)
NR0B1, (WT1mKTS | (WNT4 & CTNNB1)) & ! (NR5A1 & SOX9)
SRY, ((NR5A1 & WT1mKTS & CBX2) | (GATA4 & WT1pKTS & CBX2 & NR5A1) | (SOX9 | SRY)) & ! (CTNNB1)
SOX9, ((SOX9 & FGF9) | (SRY | PGD2) | (SRY & CBX2) | (GATA4 & NR5A1 & SRY)) & ! (WNT4 | CTNNB1 | FOXL2)
FGF9, SOX9 & ! WNT4
PGD2, SOX9
DMRT1, (SRY | SOX9) & ! (FOXL2)
DHH, SOX9
DKK1, (SRY | SOX9)
AMH, ((SOX9 & GATA4 & NR5A1) | (SOX9 & NR5A1 & GATA4 & WT1mKTS)) & ! (NR0B1 & CTNNB1)
WNT4, (GATA4 | (CTNNB1 | RSPO1 | NR0B1)) & ! (FGF9 | DKK1)
RSPO1, (WNT4 | CTNNB1) & ! (DKK1)
FOXL2, (WNT4 & CTNNB1) & ! (DMRT1 | SOX9)
CTNNB1, (WNT4 | RSPO1) & ! (SRY | (SOX9 & AMH))
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 7 of 18
Table 2 Set of regulatory interactions inferred from analysis of the dynamics of the Boolean model,
colored in orange, that deserve further experimentation to be validated
UGR, UGR & ! (NR5A1 | WNT4)
CBX2, UGR & ! (NR0B1 & WNT4 & CTNNB1)
GATA4, (UGR | WNT4 | NR5A1 | SRY)
WT1mKTS, (UGR | GATA4)
WT1pKTS, (UGR | GATA4) & ! (WNT4 & CTNNB1)
NR5A1, (UGR | CBX2 | WT1mKTS | GATA4) & ! (NR0B1 & WNT4)
NR0B1, (WT1mKTS | (WNT4 & CTNNB1)) & ! (NR5A1 & SOX9)
SRY, ((NR5A1 & WT1mKTS & CBX2) | (GATA4 & WT1pKTS & CBX2 & NR5A1) | (SOX9 | SRY)) & ! (CTNNB1)
SOX9, ((SOX9 & FGF9) | (SRY | PGD2) | (SRY & CBX2) | (GATA4 & NR5A1 & SRY)) & ! (WNT4 | CTNNB1 | FOXL2)
FGF9, SOX9 & ! WNT4
PGD2, SOX9
DMRT1, (SRY | SOX9) & ! (FOXL2)
DHH, SOX9
DKK1, (SRY | SOX9)
AMH, ((SOX9 & GATA4 & NR5A1) | (SOX9 & NR5A1 & GATA4 & WT1mKTS)) & ! (NR0B1 & CTNNB1)
WNT4, (GATA4 | (CTNNB1 | RSPO1 | NR0B1)) & ! (FGF9 | DKK1)
RSPO1, (WNT4 | CTNNB1) & ! (DKK1)
FOXL2, (WNT4 & CTNNB1) & ! (DMRT1 | SOX9)
CTNNB1, (WNT4 | RSPO1) & ! (SRY | (SOX9 & AMH))
at the beginning of simulations. Besides the wild type model, we simulated all possible
loss and gain of function of single mutants, so as to describe alterations in activation
states that might be interpreted as alterations in gene expression. Loss and gain of func-
tion single mutants were simulated by fixing the relevant node to 0 or 1, respectively. All
simulations were carried out under the synchronous updating scheme with the use of
BoolNet [33].
Testing properties of the Boolean model: random networks and robustness of attractors
We performed tests by creating random networks in order to analyze the frequency of
appearance of point attractors identical to those of the wild type model (Fig. 2). The tests
consisted in the construction of 1000 random networks with 19 nodes each one. The
number of inputs for each node in the random networks was the same as in the original
model. We kept this configuration in order to be consistent with the network architecture
of the model. The wild type attractors shown in (Fig. 2) were compared by performing
three independent tests of 1000 random networks each one. Additionally, we tested the
Fig. 2 Fixed point attractors of the Boolean model of gonadal sex determination. The attractors were
obtained by simulating all possible (219) initial activation states. The attractor with the largest basin of
attraction (50.95 %) can be interpreted as the gene expression profile observed in the somatic pre-Granulosa
cells. The attractor with the second-largest basin (48.91 %) can be interpreted as the gene expression profile
observed in the somatic pre-Sertoli cells. The model also presents a third attractor with a small basin covering
only 0.14 % of the state space interpreted as a null attractor due to a UGR node set to zero
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 8 of 18
robustness of the attractors of the BNM with a set of 1000 perturbed copies of the network
by using the testNetworkProperties function of BoolNet [33]. This test gives the percent-
age of the original attractors shown in Fig. 2 recovered after 1000 copies of the Boolean
model functions randomly perturbed.
Results
The network of gonadal sex determination
The network was constructed with 19 nodes and 78 regulatory interactions: 42 of these
interactions have been reported in humans; 12 in mice and 24 were predicted from
analysis of transition states of the simulated Boolean model. The network is directed
towards the male pathway if the SRY node is active. SRY leads to activation of SOX9,
which in turn activates FGF9, PGD2, DMRT1, DHH, DKK1, and AMH nodes. At the
same time, the female pathway is repressed by inactivating CTNNB1 and FOXL2 nodes
(Fig. 1). On the contrary, the network is directed towards the female pathway in absence
of SRY and when the WNT4, CTNNB1, RSPO1 and, FOXL2 nodes are active. In this case,
the male pathway is repressed by CTNNB1 and FOXL2 mediated inactivation of SOX9,
DMRT1 and AMH (Fig. 1).
Predictions of the Boolean model
The current model contains 24 interactions inferred from dynamic modeling, these are
predominantly related to the UGR node and the genes expressed in the bipotential
gonad. Model predictions were drawn as orange dashed lines within the following nodes:
UGR, CBX2, GATA, WT1mKTS, WT1pKTS, NR0B1, SRY, DMRT1, DKK1, WNT4 and
CTNNB1 (Fig. 1). The model predicts that the activity of UGR depends of an activation
self-loop and functions as an input to activate CBX2, GATA4, Wt1mKTS, WT1pKTS,
and NR5A1 (Table 2). The scarcity of information regarding UGR function and mainte-
nance clearly indicates that more experimental studies are necessary to understand the
mechanisms of gene expression control in the BGP especially for CBX2, GATA4 and WT1.
Dynamic behavior of the gonadal sex determination Boolean network model
The dynamic behavior of the GSD BNM was exhaustively analyzed by starting the
dynamical simulations of the system from all possible 219 = 524288 initial states. After
simulations, three fixed-point attractors where obtained (Fig. 2). The first of these attrac-
tors can be interpreted as the gene expression profile observed in Sertoli cells, the second
can be interpreted as the gene expression profile observed in granulosa cells, and the third
attractor, with a very small basin of attraction, might represent a disgenetic gonad without
Sertoli or granulosa activity.
After the initial exhaustive search of the GSD attractors, we performed a search focused
in finding the state transitions of both male and female pathways. In the case of 46,XY
simulations, the UGR node was set to ON in the initial condition (Time step 0) to transit
toward the BPG and then turning ON the SRY node leading toward the Sertoli cell attrac-
tor. Since 46,XX wild type females do not have SRY gene, we searched from all possible
initial states the activation patterns that had UGR and female nodes as initial condition.
From this search we found that UGR + WNT4 were the initial conditions (Time-step 0)
to transit from the BPG toward the granulosa attractor. Thus in male and female simula-
tions we started with an active UGR node as the initial condition, followed by activation of
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 9 of 18
the nodes representing the BPG (CBX2, GATA4, WT1mKTS, WT1pKTS, and NR5A1).
The NR0B1, WNT4 and RSPO1 nodes were subsequently activated, in agreement with
the reported gene expression patterns, showing that these genes are co-expressed in
both male and female embryos during the stage of BPG and previous to GSD (Fig. 3a
and b) [7, 34].
If the simulation transited toward the Sertoli attractor, then the NR0B1, WNT4 and
RSPO1 nodes were inactivated by NR5A1, SRY, SOX9 and DKK1 nodes. In male the
expression of NROB1 is dosage sensitive, since duplication of this gene in 46,XY patients
produces a male-to-female sex reversal with streak gonads [35]. It is important to notice
that NR0B1 might play an important role in male after the time of GSD because NR0B1
knockout mice showed disorganized Sertoli, Leydig and germ cells due to defects in
testis cord formation [36]. Thus, it has been suggested that NR0B1 has a time frame of
expression [37] with reduced levels of the DAX1 protein during GSD. Since the BNM con-
siders active or inactive states, the NR0B1 node was inactive at the sixth time step, which
corresponds to the Sertoli attractor (Fig. 3a)
When the UGR + WNT4 nodes were set to ON, two fixed point attractors were
obtained: (1) the granulosa attractor and the (2) dysgenetic gonad attractor. Thus the
transition towards the granulosa attractor is characterized by the initial activation of the
UGR + WNT4 and BPG nodes, followed by activation of the NR0B1, RSPO1, FOXL2 and
CTNNB1. As we previously stated, β-catenin, plays a key role in up-regulation of pre-
granulosa genes in female mouse [32], this factor actively antagonizes SOX9 and AMH
expression, inactivating the pathway toward Sertoli cells (Fig. 3b) [38, 39]. The attrac-
tor with no activity reflects the importance of the UGR node within the network model
a
b
Fig. 3 State transitions leading to a fixed point attractor corresponding to pre-Sertoli or to pre-granulosa
cells. The state of the nodes over time was simulated starting from an activated UGR node (1). White and
black cells represent inactive or active nodes respectively. The fixed point attractor toward pre-Sertoli is given
by activation of SRY node (a). The point attractor is in agreement with experimental observations after six
time steps, whereas the fixed point attractor toward granulosa is given by absence of SRY and activated
UGR+WNT4 nodes (b). The point attractor is in agreement with experimental observations after three time
steps
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 10 of 18
given that loss of function mutants of UGR components have an impaired subsequent
gonadal development, as observed in mouse. Therefore, the dysgenetic gonad attractor
(i.e., streaks of fibrous tissue instead of a gonad) might be interpreted as a condition
expected in some individuals when gonadal development fails, especially in the case of
LHX1, LHX9, EMX2, PAX2 and PAX8 mutants.
In summary, state transitions in Fig. 3a and b qualitatively coincide with gene expres-
sion patterns observed during GSD [7, 34]. However, notice that the state transitions
and steady state attractors must be considered as snapshots of the gene expression pat-
tern between 41–52 days of development and do not represent the complete process of
gonadal development.
Modeling disorders of sex development
46,XX sex reversal
We simulated the DSD known as 46,XX sex reversal or testicular DSD, characterized
by an apparently normal development of male structures, including testes and male
internal/external genitalia [20, 40]. To simulate such a condition either the SRY node
or the SOX9 node were left permanently active (ON = 1) during the entire simulation
(Fig. 4a, b). The SRY node activates SOX9 in coordination with CBX2, GATA4, WT1
and NR5A1, SOX9 in turn inhibits the female pathway through CTNNB1 inactivation.
CTNNB1 is the node of the transcription factor β-catenin, a key regulatory element of
the female pathway. Our BNM generates in both simulations a Sertoli-like attractor that
presents activation of the FGF9, PGD2, DMRT1, DHH, DKK1 and AMH nodes. The sim-
ulation observed in Fig. 4a might be interpreted as the process underlying a 46,XX sex
a
b
Fig. 4 Modeling 46,XX sex reversal. The SRY node was kept as active (1) so as to simulate traslocation in a
46,XX background. SRY activates SOX9 in combination with CBX2, GATA4, WT1 and NR5A1. Then, SOX9
activates nodes associated with the male pathway (i.e., FGF9, PGD2, DHH, AMH). The fixed point attractor can
be interpreted as a 46,XX sex reversal after three time steps (a). The SOX9 node was set as active in order to
simulate a duplication in a 46,XX background (b). The resulted activation states were similar to the observed
in the male pathway. Thus, the fixed point attractor can be interpreted as a 46,XX sex reversal in absence of
SRY, as reported in clinical cases
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 11 of 18
reversal when the SRY gene is translocated to one autosomic chromosome or to the X
chromosome, whereas the simulation in Fig. 4b might represent a 46,XX sex reversal due
SOX9 gene duplication.
46,XY (SRY-) sex reversal
SRY is considered the trigger of the male pathway and testis development [9]. This tran-
scription factor possesses a highly conserved domain called HMG box that binds to the
GAACAAAG DNA motif and bends the DNA molecule about 80 degrees. The loss of its
chromatin-remodeling activity [41] is considered to impair the three dimensional archi-
tecture of chromatin and compromises the proper interaction of SRY with its target genes.
Mutation of the DNA binding region of SRY in 46,XY subjects has been associated with
female external genitalia, normal Mullerian ducts and streak gonads [42]. When we sim-
ulated the SRY loss of function, the female pathway was activated by the CTNNB1 node
and the male pathway blocked through a CTNNB1 and FOXL2- mediated SOX9 inhi-
bition. SRY is considered the trigger of testis development by expressing in the somatic
pre-Sertoli cells [9]. The mechanism suggested for normal function of this transcrip-
tion factor is a highly conserved domain within the protein, called High Mobility Group
(HMG box).
Concerning the model simulation in Fig. 5c, loss-of-function of SRY leads to inacti-
vation of the male and activation of the female pathway. To this respect, Hawkings and
colleagues [42] described five subjects with 46,XY karyotype associated with completely
female external genitalia, normal Mullerian ducts, and streak gonads. All the patients
showed mutations in the DNA binding region of the SRY protein [42]. Since model sim-
ulations agree with clinical observations of loss-of-function mutations in the HMG box
of SRY, we interpret this simulation as the possible gene expression dynamics in a 46,XY
(SRY-) individual during the time of GSD. In these subjects the female pathway would
become active by increasing amounts of β-catenin within the cell nucleus and active
repression of the male pathway by a B-catenin and FOXL2-mediated inhibition of SOX9.
Given this results we interpret that our simulations (Fig. 5c) resemble the early gene
expression dynamics in a 46,XY (SRY-) individuals.
Modeling other DSDs
Other relevant elements that have a common function in both male and female pathways
at the BPG and thorough the differentiation of testis and ovaries are the transcription
factors GATA4 and WT1. In the case of GATA4, it has been observed in mice that
GATA4 is expressed at E10.5 during formation of the UGR and its deficiency impairs
subsequent gonadal differentiation [43]. In the male pathway, GATA4 associates with
the -KTS isoform of WT1 protein for an optimal activation of the SRY gene [44]. Other
example of the GATA4 protein activity in the male pathway is its role in the activa-
tion of the AMH gene in association with SF1 and WT1-KTS transcription factors
[45, 46]. Simulation of GATA4 loss of function in the male pathway is given in Fig. 6
(notice the altered dynamics of activation patterns compared with the wild-type simula-
tion shown in Fig. 3a). WT1mKTS, WT1pKTS, NR5A1 and AMH nodes were inactive
in the attractor because the GATA4 node is their positive regulator. The SRY node
remained active due to an activation self-loop and additional interactions with NR5A1,
WT1mKTS, CBX2 and a possible feedback loop with SOX9, thus the altered activation
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 12 of 18
Fig. 5 Modeling 46,XY sex reversal. The CBX2 node was inactivated in order to simulate a loss of function
mutation in a 46,XY background which resulted in a steady state attractor that showed activation of female
nodes (a). The NR0B1 node was set as active in order to simulate a duplication in a 46,XY background. The
activation states were altered and resulted in an steady state attractor with activated female nodes (b). The
SRY node was inactivated in order to simulate a loss of function mutation which resulted in a steady state
attractor that showed activation of female nodes (c)
Fig. 6 State transitions when the GATA4 node was inactive (0) in a 46,XY context. The GATA4 node was
inactivated in order to simulate a loss of function mutation in a 46,XY background which resulted in
inactivated AMH node although an attractor with activated male nodes was recovered
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 13 of 18
state shown in Fig. 6 might be interpreted as the source of a DSD. To this respect, the
clinical spectrum of developmental anomalies due to GATA4 mutations in male patients
is variable. Patients might show bilateral dysgenetic testes containing Sertoli cells and
no visible Leydig cells and show male internal genitalia to normal-ambiguous external
genitalia [46].
On the other hand, WT1 has a key role in the development of kidney and gonads, its
expression is observed at the UGR and continues through the differentiation of testis
and ovaries interacting in both pathways of cell differentiation. Homozygous mutations
in mice are embryonic lethal and result in renal agenesis, as well as cardiac and gen-
ital tract abnormalities [29]. Since WT1 is an important element in early stages of
gonadal development we simulated the loss of function of WT1 in a 46,XY context
(Fig. 7), notice that the attractor has an activation pattern similar to the female path-
way. Importantly, mutations in this gene impair the development of male in a certain
degree; mutations in the intron 9 splice site of the WT1 gene affects the balance of
the +KTS/-KTS isoforms impairing the development of testis resulting in streak gonads
and ambiguous external genitalia [47]. In females the role of WT1 is less defined, how-
ever there is experimental evidence about its role in the positive regulation of the
NR0B1 gene by binding to two potential sites located at the 5 flanking region of the
gene [48].
We analyzed in addition the effect of loss-of-function mutations in the NR5A1 node.
Since the dynamics of the simulations of WT1 and NR5A1 were identical we show only
the attractor in Fig. 7.
Testing properties of the Boolean model: random networks and robustness of the attractors
On average, the three independent tests of 1000 random networks generated 103,000
attractors each one. We found that none of the random networks recovered the set of
three point attractors of the wild type model shown in Fig. 2. This results indicate that the
attractors of our BNM could not be expected in a network made with random interac-
tions. Thus, the attractors in Fig. 2 can be considered as biologically meaningful and not
a statistical artifact.
Concerning the test of attractor robustness we observed that 95 % of the perturbed
networks recovered less than 30 % of the original attractors of our model. This means
that the model is relatively sensitive to perturbations in the functions of the model. The
reduced robustness can be due to the scarce redundancy in the model, given that we
opted by including a small number of regulatory molecules, so as to be close to a minimal
model. We expect that the introduction of more nodes and regulatory feedback circuits
would result in an increased robustness.
Fig. 7 Fixed point attractors when setting NR5A1 or WT1 inactive (0) in a 46,XY context
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 14 of 18
Discussion
The classical observations of Alfred Jost (1947) that early castration in utero of rabbit
fetuses resulted in female internal and external genitalia (independent of their chro-
mosomal sex complement) lead to the hypothesis of a testis-determining factor (TDF).
According to this hypothesis the ovary was considered the default developmental state,
while testis represented an induced and active state that repressed female development.
Experimental evidence accumulated in the last 20 years have enriched our view of
sex determination where developmental programs toward testis or ovaries represent
two independent antagonistic regulatory pathways of high complexity intertwined in a
regulatory network: the GSD network.
The BNM used in this study, although discrete in its approach, can be considered as a
simplified version of a very dynamic and complex biological network that incorporates
the major regulatory elements of the GSD network. The GSD BNM summarizes in a
formal language the set of experimentally-confirmed interactions associated with the pro-
cess of GSD. Although the attractors obtained in our model cannot be interpreted as as
anatomical structures of high developmental complexity, they can be reliably seen as the
gene expression profiles expected during the process of determination and differentiation
of SPC and GPC towards Sertoli and granulosa cells respectively.
The network of gonadal sex determination
We inferred the regulatory network of human GSD and modeled it as a BNM. With such a
model we were able to describe the molecular dynamics of the first stage in gonadal mor-
phogenesis, which is the cell fate determination and further differentiation of Sertoli and
granulosa cells. The network contains 19 nodes as well as their regulatory interactions, as
evidenced by published experimental and clinical data.
Recent studies regarding early gonadal differentiation suggest a highly complex biolog-
ical process regulated by many, probably hundreds, of genes. However, our BNM shows
that only a handful of them are sufficient to activate the male or female pathway, allowing
us to propose that the gonadal fate commitment and differentiation is a direct conse-
quence of activation and repression of a transcriptional program encoded as a regulatory
network. Although part of the information used to infer this regulatory network was taken
from experiments in mice, we consider that the model might be considered as a good
approximation to the corresponding regulatory network in humans.
The Boolean model of gonadal sex determination
The BNM describes the dynamics of 19 nodes associated with GSD between 41–52 days
of embryonic development. The results cannot be considered as final activation states
of the biological process, instead they should be considered as a snapshot in the pro-
cess of Sertoli and granulosa cell differentiation. At the quantitative level, GSD is poorly
understood given the lack of information about kinetic details of each regulatory ele-
ment, therefore it is difficult to establish a continuous model with differential equations
with the current available data. Despite the large amount of gene expression data, little
is known about the regulatory mechanisms leading to GSD under normal conditions, as
well as their downstream effects under mutant conditions. To shed light about the regula-
tory mechanisms we used a discrete modeling approach because most of the information
relies on qualitative descriptions.
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 15 of 18
Model predictions and the role of the UGR and bipotential gonad genes in early gonadal
development
The UGR is a very important structure within the developing embryo since it is the com-
mon structure that leads to testis and ovaries. Notice that just a few regulatory elements
of the UGR have been studied. For example, Lhx1 expression has been reported in mice
and has a key role in the development of kidney, female reproductive tract and anterior
head [30]. Conditional knockout mice lack uterus, cervix and upper vagina [49]. Lhx9 has
a role in the activation of Nr5a1 gene, in synergy with Wt1 in mice. Other example is
given by PAX2 and PAX8 genes. In humans, these genes have a role in the activation of
the WT1 gene [29, 50] thus, we underline the need for additional studies regarding the
regulatory interactions that led to the establishment of the UGR and BPG primordium.
As we mentioned previously, the human female pathway is less characterized, and its
current cumulative knowledge is mainly based on mice findings. In this case, the GATA4-
FOG2 complex has an important function by activating the Fst, Wnt4, Sprr2d, Foxl2,
Gng13 genes [32]. It has been observed in female mice that loss of function of Gata4
impairs the expression of these genes and leads to the development of a male-specific
coelomic vessel [32]; therefore GATA4 can be considered as a key regulatory element in
the early stages of gonadal development toward ovaries.
The role of WNT4/β-catenin in the female pathway
Concerning the female pathway most of the inferred interactions derive from mice. To
this respect we notice the role of the WNT4 and βcatenin nodes in the regulation of
WNT4, RSPO1, and FOXL2. In the biological process we predict a key role of β-catenin
regulating the female pathway. The general mechanism of the canonical Wnt4/β-catenin
signaling pathway can be explained as follows: the pathway is initiated by expression of
the wingless-type MMTV integration site family, member 4 (WNT4). The product of this
gene is a ligand that binds to Frizzled (Fz) and LRP5/6 co-receptors at the plasma mem-
brane, disengaging β-catenin from the proteins of the “destruction complex”? (Axin and
APC). Then β-catenin translocate into the nucleus where associates with TCF7/LEF, this
protein contains an HMG box with capacity to recognize specific DNA sequences. The
β-catenin-TCF7/LEF complex activates target genes, whereas in absence of β-catenin
TCF7/LEF alone represses gene transcription [51, 52]. From the set of interactions
inferred from mice, that fitted perfectly in our model, we would expect that β-catenin
might have an important role regulating the expression of the FST, FOXL2 and IRX3 genes
in humans.
The BNM of GSD summarizes in a formal language the set of experimentally-confirmed
interactions associated with the process of GSD. The attractors of our model can be
interpreted as the gene expression profiles expected during the process of GSD and dif-
ferentiation of Sertoli or granulosa cell lineages. According to our simulations, the loss of
function of GATA4 results in inactivation of the AMH node in the attractor. This result
is particularly interesting given the existence of the persistent Müllerian duct syndrome
(PMDS), a relatively rare inherited defect in the sexual differentiation, characterized by
failure in the regression of the Müllerian ducts in males. Affected individuals present per-
sistent uterus and tubes due to a defect in the synthesis of the AMH hormone, which is
normally produced by the Sertoli cells. Mutations in the AMH gene have been reported
in these patients [20, 53], however the majority of PMDS remain without molecular
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 16 of 18
diagnosis, therefore GATA4 mutations emerge, according to our model predictions, as a
potential PMSD causing gene.
Conclusions
We propose the present model as a starting point for future mathematical modeling
and integration of experimental research regarding sex development. The model can be
upgraded in several aspects for example, incorporating additional nodes and interactions,
as well as modeling more cell lineages of the gonad such as the Leydig or theca cells.
Finally the current BNM describes the dynamics of the GSD network under perturba-
tions. Importantly the analysis of these states can have potential implications in the study
of DSDs.
Additional file
Additional file 1: Supplementary information. (PDF 47 kb)
Competing interests
The authors declare that they have no competing interests.
Authors’ contributions
LT and SF conceived the project; OR, LM and AR developed the BNM and performed the simulations; LT and LM
coordinated computational work; OR, LT, LM, AR and SF, analyzed the data.; SK and HM provided important contribution
for including nodes and interactions and for discussion; OR, SF, and LT, wrote the manuscript. All authors read and
approved the final manuscript.
Acknowledgements
OR thanks the support from the program Doctorado en Ciencias Biológicas, UNAM, and the scholarship 173000 from
Consejo Nacional de Ciencia y Tecnología (CONACyT). This work was supported by grants from CONACYT 166012 to HM,
Universidad Nacional Autónoma de México PAPIIT IN200514 to LM, and Recursos Fiscales para la Investigación del
Instituto Nacional de Pediatría, proyecto 057/2014 to LT. Adhemar Liquitaya from CompBioLab IIB, UNAM, aid with the
test of random networks.
Author details
1Instituto Nacional de Pediatría, Laboratorio de Citogenética, Av. Insurgentes Sur 3700 C, 04530 México City, México.
2Programa de Doctorado en Ciencias Biológicas, UNAM, Mexico City, México. 3Instituto de Investigaciones Biomédicas,
UNAM, 04510 Mexico City, México. 4Programa de Doctorado en Ciencias Biomédicas, UNAM, Mexico City, México.
5Facultad de Medicina/Hospital General de Mexico, Mexico City, México. 6C3, Centro de Ciencias de la Complejidad,
UNAM, 04510 Mexico City, México.
Received: 21 August 2015 Accepted: 30 October 2015
References
1.
De Santa Barbara P, Moniot B, Poulat F, Berta P. Expression and subcellular localization of SF-1, SOX9, WT1, and
AMH proteins during early human testicular development. Dev Dynam. 2000;217(3):293–8.
2.
Eggers S, Sinclair A. Mammalian sex determination insights from humans and mice. Chromosome Res. 2012;20(1):
215–38.
3.
White S, Sinclair A. The molecular basis of gonadal development and disorders of sex development In: Hutson JM,
Warne GL, Grover SR, editors. Disorders of sex development: An Integrated Approach to Management. Heidelberg:
Springer Berlin; 2012. p. 1–9.
4.
She ZY, Yang WX. Molecular mechanisms involved in mammalian primary sex determination. J Mol Endocrinol.
2014;53(1):R21–37.
5.
Sajjad Y. Development of the genital ducts and external genitalia in the early human embryo. J Obstet Gynaecol Re.
2010;36(5):929–37.
6.
Hutson JM. Embryology of the human genital tract In: Hutson JM, Warne GL, Grover SR, editors. Disorders of sex
development: An Integrated Approach to Management. Heidelberg: Springer Berlin; 2012. p. 11–21.
7.
Hanley NA, Hagan DM, Clement-Jones M, Ball SG, Strachan T, Salas-Cortes L, et al. SRY, SOX9, DAX1 expression
patterns during human sex determination and gonadal development. Mech Develop. 2000;91(1):403–7.
8.
Kim Y, Capel B. Balancing the bipotential gonad between alternative organ fates: a new perspective on an old
problem. Dev Dynam. 2006;235(9):2292–300.
9.
Albrecht KH, Eicher EM. Evidence that SRY is expressed in pre-Sertoli cells and Sertoli and granulosa cells have a
common precursor. Dev Biol. 2001;240(1):92–107.
10. Ungewitter EK, Yao HC. How to make a gonad: cellular mechanisms governing formation of the testes and ovaries.
Sex Dev. 20013;7(1-3):7–20.
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 17 of 18
11. Kashimada K, Koopman P. SRY: the master switch in mammalian sex determination. Development. 2010;137(23):
3921–30.
12. Harikae K, Miura K, Kanai Y. Early gonadogenesis in mammals: significance of long and narrow gonadal structure.
Dev Dynam. 2013;242(4):330–8.
13. Sarraj MA, Drummond AE. Mammalian foetal ovarian development: consequences for health and disease.
Reproduction. 2012;143(2):151–63.
14. Svechnikov K, Landreh L, Weisser J, Izzo G, Colon E, Svechnikova I, et al. Origin development and regulation of
human Leydig cells. Horm Res Paediatr. 2010;73(2):93–101.
15. Beverdam A, Koopman P. Expression profiling of purified mouse gonadal somatic cells during the critical time
window of sex determination reveals novel candidate genes for human sexual dysgenesis syndromes. Hum Mol
Genet. 2006;15(3):417–31.
16. Jameson SA, Natarajan A, Cool J, DeFalco T, Maatouk DM, Mork L, et al. Temporal transcriptional profiling of
somatic and germ cells reveals biased lineage priming of sexual fate in the fetal mouse gonad. PLoS Genet.
2012;8(3):e1002575.
17. Munger SC, Natarajan A, Looger LL, Ohler U, Capel B. Fine time course expression analysis identifies cascades of
activation and repression and maps a putative regulator of mammalian sex determination. PLoS Genet. 2013;9(7):
e1003630.
18. Hughes IA, Houk Ch, Ahmed SF, Lee PA. Consensus statement on management of intersex disorders. J Pediatr Urol.
2006;2(3):148–62.
19. Mendonca BB, Domenice S, Arnhold IJ, Costa EM. 46,XY disorders of sex development (DSD). Clin Endocrinol.
2009;70(2):173–87.
20. López M, Torres L, Méndez JP, Cervantes A, Perez-Palacios G, Erickson RP. Clinical traits and molecular finding in
46,XX males. Clin Genet. 1995;48(1):29–34.
21. Kousta E, Papathanasiou A, Skordis N. Sex determination and disorders of sex development according to the
revised nomenclature and classification in 46, XX individuals. Hormones (Athens). 2010;9(3):218–31.
22. Saadatpour A, Albert R. Boolean modeling of biological regulatory networks: a methodology tutorial. Methods.
2013;62(1):3–12.
23. Karlebach G, Shamir R. Modelling, analysis of gene regulatory networks. Nat Rev Mol Cell Bio. 2008;9(10):770–80.
24. Sanchez-Corrales YE, Alvarez-Buylla ER, Mendoza L. The Arabidopsis thaliana flower organ specification gene
regulatory network determines a robust differentiation process. J Theor Biol. 2010;264(3):971–83.
25. Herrmann F, Groß A, Zhou D, Kestler HA, Kühl M. A Boolean model of the cardiac gene regulatory network
determining first and second heart field identity. PLoS ONE. 2012;7(10):e46798.
26. Albert R, Othmer HG. The topology of the regulatory interactions predicts the expression pattern of the segment
polarity genes in Drosophila melanogaster. J Theor Biol. 2003;223(1):1–18.
27. Fleming A, Vilain E. The endless quest for sex determination genes. Clin genet. 2005;67(1):15–25.
28. Ono M, Harley VR. Disorders of sex development: new genes new concepts. Nat Rev Endocrinol. 2013;9(2):79–91.
29. McConnell MJ, Cunliffe HE, Chua LJ, Ward TA, Eccles MR. Differential regulation of the human Wilms tumour
suppressor gene (WT1) promoter by two isoforms of PAX2. Oncogene. 1997;14(22):2689–700.
30. Kobayashi A, Shawlot W, Kania A, Behringer RR. Requirement of Lim1 for female reproductive tract development.
Development. 2004;131(3):539–49.
31. Wilhelm D, Englert Ch. The Wilms tumor suppressor WT1 regulates early gonad development by activation of SF1.
Genes Dev. 2002;16(14):1839–51.
32. Manuylov NL, Smagulova FO, Leach L, Tevosian SG. Ovarian development in mice requires the GATA4-FOG2
transcription complex. Development. 2008;135(22):3731–43.
33. Müssel Ch, Hopfensitz M, Kestler HA. BoolNet an R package for generation reconstruction and analysis of Boolean
networks. Bioinformatics. 2010;26(10):1378–80.
34. Tomaselli S, Megiorni F, Lin L, Mazzilli MC, Gerrelli D, Majore S, et al. Human RSPO1/R-spondin1 is expressed
during early ovary development and augments β-catenin signaling. PLoS One. 2011;6(1):e16366.
35. Barbaro M, Oscarson M, Schoumans J, Staaf J, Ivarsson SA, Wedell A. Isolated 46, XY gonadal dysgenesis in two
sisters caused by a Xp21. 2 interstitial duplication containing the DAX1 gene. J Clin Endocrinol Metab. 2007;92(8):
3305–13.
36. Meeks JJ, Crawford SE, Russell TA, Morohashi K, Weiss J, Jameson JL. Dax1 regulates testis cord organization
during gonadal differentiation. Development. 2003;130(5):1029–36.
37. Ludbrook LM, Harley VR. Sex determination: a window of DAX1 activity. Trends Endocrin Met. 2004;15(3):116–21.
38. Hersmus R, Kalfa N, de Leeuw BHCGM, Stoop H, Oosterhuis JW, de Krijger R, et al. FOXL2 and SOX9 as parameters
of female and male gonadal differentiation in patients with various forms of disorders of sex development (DSD). J
Pathol. 2008;215(1):31–8.
39. Maatouk DM, DiNapoli L, Alvers A, Parker KL, Taketo MM, Capel B. Stabilization of β-catenin in XY gonads causes
male-to-female sex-reversal. Hum Mol Genet. 2008;17(19):2949–55.
40. Alves C, Braid Z, Coeli FB, Mello MP. 46 XX Male-testicular disorder of sexual differentiation (DSD): hormonal
molecular and cytogenetic studies. Arq Bras Endocrinol. 2010;54(8):685–9.
41. Harley VR, Clarkson MJ, Argentaro A. The molecular action and regulation of the testis-determining factors SRY
(Sex-determining Region on the Y chromosome) and SOX9 [SRY-related high-mobility group (HMG) box 9]. Endocr
rev. 2003;24(4):466–87.
42. Hawkins JR, Taylor A, Goodfellow PN, Migeon CJ, Smith KD, Berkovitz GD. Evidence for increased prevalence of SRY
mutations in XY females with complete rather than partial gonadal dysgenesis. Am J Hum Genet. 1992;51(5):979.
43. Yueh-Chiang Hu, Okumura LM, Page DC. Gata4 is required for formation of the genital ridge in mice. PLoS Genet.
2013;9(7):e1003629.
44. Miyamoto Y, Taniguchi H, Hamel F, Silversides DW, Viger RS. A GATA4/WT1 cooperation regulates transcription of
genes required for mammalian sex determination and differentiation. BMC Mol Biol. 2008;9(1):44.
Ríos et al. Theoretical Biology and Medical Modelling (2015) 12:26
Page 18 of 18
45. Tremblay JJ, Viger RS. A mutated form of Steroidogenic factor 1 (SF-1 G35E that causes sex reversal in humans fails
to synergize with transcription factor GATA-4. J Biol Chem. 2003;278(43):42637–42.
46. Lourenço D, Brauner R, Rybczy´nska M, Nihoul-Fékété C, McElreavey K, Bashamboo A. Loss-of-function mutation in
GATA4 causes anomalies of human testicular development. Proc Natl Acad Sci U S A. 2011;108(4):1597–602.
47. Köhler B, Biebermann H, Friedsam V, Gellermann J, Maier RF, Pohl M, et al. Analysis of the Wilms’ tumor suppressor
gene (WT1) in patients 46, XY disorders of sex development. Clin Endocrinol Metab. 2011;96(7):E1131–6.
48. Kim J, Prawitt D, Bardeesy N, Torban E, Vicaner C, Goodyer P, et al. The Wilms’ tumor suppressor gene WT1
product regulates DAX-1 gene expression during gonadal differentiation. Mol Cellular Biol. 1999;19(3):2289–99.
49. Huang C-C, Orvis GD, Kwan KM, Behringer RR. Lhx1 is required in Müllerian duct epithelium for uterine
development. Dev Biol. 2014;389(2):124–36.
50. Fraizer GC, Shimamura R, Zhang X, Saunders GF. PAX8 regulates human WT1 transcription through a novel DNA
binding site. J Biol Chem. 1997;272(49):30678–87.
51. Cadigan KM, Waterman ML. TCF/LEFs and Wnt signaling in the nucleus. Cold Spring Harbor Perspectives Biol.
2012;4(11):a007906.
52. Tevosian SG, Manuylov NL. To β or not to β Canonical β-catenin signaling pathway and ovarian development. Dev
Dynam. 2008;237(12):3672–80.
53. Mazen I, Hamid A, El-Gammal M, Aref A, Amr K. AMH gene mutations in two Egyptian families with persistent
Müllerian duct syndrome. Sex Dev. 2011;5(6):277–80.
Submit your next manuscript to BioMed Central
and take full advantage of:
• Convenient online submission
• Thorough peer review
• No space constraints or color figure charges
• Immediate publication on acceptance
• Inclusion in PubMed, CAS, Scopus and Google Scholar
• Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
|
26573569
|
WT1mKTS = ( GATA4 ) OR ( UGR )
CTNNB1 = ( ( ( RSPO1 ) AND NOT ( SRY ) ) AND NOT ( SOX9 AND ( ( ( AMH ) ) ) ) ) OR ( ( ( WNT4 ) AND NOT ( SRY ) ) AND NOT ( SOX9 AND ( ( ( AMH ) ) ) ) )
AMH = ( ( SOX9 AND ( ( ( GATA4 AND NR5A1 ) ) OR ( ( WT1mKTS AND GATA4 AND NR5A1 ) ) ) ) AND NOT ( NR0B1 AND ( ( ( CTNNB1 ) ) ) ) )
UGR = ( ( ( UGR ) AND NOT ( WNT4 ) ) AND NOT ( NR5A1 ) )
NR5A1 = ( ( UGR ) AND NOT ( NR0B1 AND ( ( ( WNT4 ) ) ) ) ) OR ( ( GATA4 ) AND NOT ( NR0B1 AND ( ( ( WNT4 ) ) ) ) ) OR ( ( CBX2 ) AND NOT ( NR0B1 AND ( ( ( WNT4 ) ) ) ) ) OR ( ( WT1mKTS ) AND NOT ( NR0B1 AND ( ( ( WNT4 ) ) ) ) )
DHH = ( SOX9 )
FGF9 = ( ( SOX9 ) AND NOT ( WNT4 ) )
SOX9 = ( ( ( ( PGD2 ) AND NOT ( CTNNB1 ) ) AND NOT ( FOXL2 ) ) AND NOT ( WNT4 ) ) OR ( ( ( ( GATA4 AND ( ( ( SRY AND NR5A1 ) ) ) ) AND NOT ( CTNNB1 ) ) AND NOT ( FOXL2 ) ) AND NOT ( WNT4 ) ) OR ( ( ( ( SRY ) AND NOT ( CTNNB1 ) ) AND NOT ( FOXL2 ) ) AND NOT ( WNT4 ) ) OR ( ( ( ( FGF9 AND ( ( ( SOX9 ) ) ) ) AND NOT ( CTNNB1 ) ) AND NOT ( FOXL2 ) ) AND NOT ( WNT4 ) ) OR ( ( ( ( CBX2 AND ( ( ( SRY ) ) ) ) AND NOT ( CTNNB1 ) ) AND NOT ( FOXL2 ) ) AND NOT ( WNT4 ) )
WT1pKTS = ( ( GATA4 ) AND NOT ( WNT4 AND ( ( ( CTNNB1 ) ) ) ) ) OR ( ( UGR ) AND NOT ( WNT4 AND ( ( ( CTNNB1 ) ) ) ) )
WNT4 = ( ( ( CTNNB1 ) AND NOT ( FGF9 ) ) AND NOT ( DKK1 ) ) OR ( ( ( NR0B1 ) AND NOT ( FGF9 ) ) AND NOT ( DKK1 ) ) OR ( ( ( GATA4 ) AND NOT ( FGF9 ) ) AND NOT ( DKK1 ) ) OR ( ( ( RSPO1 ) AND NOT ( FGF9 ) ) AND NOT ( DKK1 ) )
FOXL2 = ( ( ( WNT4 AND ( ( ( CTNNB1 ) ) ) ) AND NOT ( SOX9 ) ) AND NOT ( DMRT1 ) )
PGD2 = ( SOX9 )
DMRT1 = ( ( SOX9 ) AND NOT ( FOXL2 ) ) OR ( ( SRY ) AND NOT ( FOXL2 ) )
GATA4 = ( NR5A1 ) OR ( UGR ) OR ( WNT4 ) OR ( SRY )
DKK1 = ( SRY ) OR ( SOX9 )
NR0B1 = ( ( WNT4 AND ( ( ( CTNNB1 ) ) ) ) AND NOT ( NR5A1 AND ( ( ( SOX9 ) ) ) ) ) OR ( ( WT1mKTS ) AND NOT ( NR5A1 AND ( ( ( SOX9 ) ) ) ) )
SRY = ( ( CBX2 AND ( ( ( WT1mKTS AND NR5A1 ) ) ) ) AND NOT ( CTNNB1 ) ) OR ( ( GATA4 AND ( ( ( CBX2 AND WT1pKTS AND NR5A1 ) ) ) ) AND NOT ( CTNNB1 ) ) OR ( ( SOX9 ) AND NOT ( CTNNB1 ) ) OR ( ( SRY ) AND NOT ( CTNNB1 ) )
CBX2 = ( ( UGR ) AND NOT ( NR0B1 AND ( ( ( WNT4 AND CTNNB1 ) ) ) ) )
RSPO1 = ( ( WNT4 ) AND NOT ( DKK1 ) ) OR ( ( CTNNB1 ) AND NOT ( DKK1 ) )
|
RESEARCH ARTICLE
A Network Model to Describe the Terminal
Differentiation of B Cells
Akram Méndez1,2, Luis Mendoza2,3*
1 Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Ciudad de
México, México, 2 Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México,
Ciudad de México, México, 3 C3, Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de
México, Ciudad de México, México
* lmendoza@biomedicas.unam.mx
Abstract
Terminal differentiation of B cells is an essential process for the humoral immune response
in vertebrates and is achieved by the concerted action of several transcription factors in
response to antigen recognition and extracellular signals provided by T-helper cells. While
there is a wealth of experimental data regarding the molecular and cellular signals involved
in this process, there is no general consensus regarding the structure and dynamical prop-
erties of the underlying regulatory network controlling this process. We developed a dynam-
ical model of the regulatory network controlling terminal differentiation of B cells. The
structure of the network was inferred from experimental data available in the literature, and
its dynamical behavior was analyzed by modeling the network both as a discrete and a con-
tinuous dynamical systems. The steady states of these models are consistent with the pat-
terns of activation reported for the Naive, GC, Mem, and PC cell types. Moreover, the
models are able to describe the patterns of differentiation from the precursor Naive to any of
the GC, Mem, or PC cell types in response to a specific set of extracellular signals. We sim-
ulated all possible single loss- and gain-of-function mutants, corroborating the importance
of Pax5, Bcl6, Bach2, Irf4, and Blimp1 as key regulators of B cell differentiation process.
The model is able to represent the directional nature of terminal B cell differentiation and
qualitatively describes key differentiation events from a precursor cell to terminally differenti-
ated B cells.
Author Summary
Generation of antibody-producing cells through terminal B cell differentiation represents
a good model to study the formation of multiple effector cells from a progenitor cell type.
This process is controlled by the action of several molecules that maintain cell type specific
programs in response to cytokines, antigen recognition and the direct contact with T
helper cells, forming a complex regulatory network. While there is a large body of experi-
mental data regarding some of the key molecules involved in this process and there have
been several efforts to reconstruct the underlying regulatory network, a general consensus
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
1 / 26
a11111
OPEN ACCESS
Citation: Méndez A, Mendoza L (2016) A Network
Model to Describe the Terminal Differentiation of B
Cells. PLoS Comput Biol 12(1): e1004696.
doi:10.1371/journal.pcbi.1004696
Editor: Thomas Höfer, German Cancer Research
Center, GERMANY
Received: June 17, 2015
Accepted: December 7, 2015
Published: January 11, 2016
Copyright: © 2016 Méndez, Mendoza. This is an
open access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Funding: This work was funded by Programa de
Apoyo a Proyectos de Investigación e Inovación
Tecnológica, grant IN200514 to LM, Dirección
General de Asuntos del Personal Académico de la
Universidad Nacional Autónoma de México (http://
dgapa.unam.mx); and Scholarship 384318 to AM,
Consejo Nacional de Ciencia y Tecnología (http://
www.conacyt.mx). The funders had no role in study
design, data collection and analysis, decision to
publish, or preparation of the manuscript.
about the structure and dynamical behavior of this network is lacking. Moreover, it is not
well understood how this network controls the establishment of specific B cell expression
patterns and how it responds to specific external signals. We present a model of the regula-
tory network controlling terminal B cell differentiation and analyze its dynamical behavior
under normal and mutant conditions. The model recovers the patterns of differentiation
of B cells and describes a large set of gain- and loss-of-function mutants. This model pro-
vides an unified framework to generate qualitative descriptions to interpret the role of
intra- and extracellular regulators of B cell differentiation.
Introduction
Adaptive immunity in vertebrates depends on the rapid maturation and differentiation of T
and B cells. While T cells originate cell-mediated immune responses, B cells are responsible for
the humoral response of the organism by means of the production of high-affinity antibodies.
B cells develop in the bone marrow from hematopoietic progenitors, and migrate as mature B
cells (Naive) to the germinal centers (GCs), which are highly specialized environments of the
secondary lymphoid organs [1]. There, B cells are activated by antigens (Ag) and undergo
diversification of the B cell receptor (BCR) genes by somatic hypermutation (SHM), as well as
the subsequent expression of distinct isotypes by class switch recombination (CSR) [2]. After
the activation due to Ag recognition, Naive and GC B cells differentiate into antibody-produc-
ing plasma cells (PC), as well as memory cells (Mem) [3]. Cytokines secreted by T-helper cells,
such as IL-2, IL-4 and IL-21 as well as the direct contact with these cells, mediated by the union
CD40 receptor on B cells with its ligand CD40L, play a key role in the determination of B cell
fate [4], since these external signals act as instructive cues that promote the differentiation
from a cell progenitor to multiple cell types (Fig 1).
Terminal differentiation of B cells is controlled by the concerted action of multiple tran-
scription factors that integrate physiologic signals in response to BCR cross-linking, extracellu-
lar cytokines, and the direct interaction with T cells, thus creating a complex regulatory
network. These factors appear to regulate mutually antagonistic programs and can be divided
into those that promote and maintain B cell identity, such as Pax5, Bcl6 and Bach2, and those
that control differentiation into memory cells or plasma cells, i.e., Irf4, Blimp1 and XBP1, as
has been shown by multiple functional, biochemical and gene expression analysis [5–7].
A type is characterized by the expression of a specific set of master transcriptional regula-
tors. Naive B cells express Pax5 and Bach2, which are induced at the onset of B cell develop-
ment, and are maintained through all developmental stages upon plasma cell differentiation [8,
9]. Furthermore, Pax5 is essential for the maintenance of B cell identity, since Pax5 deficiency
results in the acquisition of multilineage potential [10]. Both Pax5 and Bach2 are required to
inhibit PC differentiation [11, 12]. In addition to Pax5 and Bach2, GC cells express Bcl6, a tran-
scription factor necessary for germinal center formation that allows the SHM and CSR pro-
cesses to occur [13–15]. Development of B cells toward Mem cells requires Bcl6
downregulation and the induction of Irf4 [16, 17]. Conversely, PCs are characterized by the
expression of Blimp1 and XBP1 that along with Irf4, inhibit the B cell identity program [5, 18].
Although a number of molecules that play a key role in the process of the terminal differen-
tiation of B cells are known, it is not completely clear how such molecules regulate each other
to ensure the proper appearance of GC, Mem, and PC from progenitor Naive B cells. There
exist models describing several aspects of the differentiation of B cells such as the decisions pro-
moting the developmental processes of CSR and SHM [19, 20], the response to environmental
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
2 / 26
Competing Interests: The authors have declared
that no competing interests exist.
contaminants that disrupt B cell differentiation [21, 22], the B cell exit from the GC phase for
the differentiation into plasma or memory cells [23], as well as the dynamics of B cell differenti-
ation inside the complex microenvironment of germinal centers [24, 25]. Nonetheless, a gen-
eral consensus about the regulatory network controlling cell fate decisions of B lymphocytes is
lacking.
The modeling of regulatory networks has been shown to be a valuable approach to under-
stand the way cells integrate several signals that control the differentiation process [26, 27]. In
particular, the logical modeling approach has been useful to qualitatively describe biological
processes for which detailed kinetic information is lacking [28]. This type of modeling usually
focus on the nature and number of steady states reached by the network, which are often inter-
preted as stable patterns of gene expression that characterize multiple cell fates [29]. In this par-
adigm, the transit from one steady state to another occurs when cells receive a specific external
stimuli, such as hormones, cytokines, changes in osmolarity, etc. These external stimuli are
sensed and integrated to create an intracellular response that may trigger a global response
such as cell growth, division, differentiation, etc. External signals are usually continuous in
nature, i.e., they are present as concentration gradients of external molecules that may attain
different values of strength and duration. Therefore it becomes desirable to develop models
Fig 1. Terminal B cell differentiation. Precursor Naive B cells can differentiate into three possible cell types
depending on proper molecular stimuli. Cytokines secreted by T-helper cells play a central role in the
determination of B cell fate. IL-2 and IL-4 are required for the transition of Naive to GC cells. Direct contact of
B cells with T cells by means of the CD40L receptor promote the differentiation of Naive or GC cells toward
the Mem cell type. Antigen (Ag) activation drives terminal differentiation toward the PC cells, a process that is
favored by the presence of IL-21.
doi:10.1371/journal.pcbi.1004696.g001
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
3 / 26
that incorporate the possibility of following the response of the network to continuous signals
while at the same time, describe qualitatively the directional and branched nature of cell differ-
entiation processes.
In this work we infer the regulatory network that controls the terminal differentiation of B
cells. We then construct two dynamical systems, one discrete and one continuous, to analyze
the dynamical properties of the regulatory network. Specifically, we find the stationary states of
the models, and compare them against the known stationary molecular patterns observed in
Naive, GC, Mem, and PC cells, under wild type and mutant backgrounds. Finally, we show
that the dynamical models are able to describe the cellular differentiation pattern under a vari-
ety of external signals. Importantly, the models predict the existence of several interactions nec-
essary for the network to ensure the proper pattern of terminal differentiation of B cells.
Furthermore, the continuous model predicts the existence of intermediary states that could be
reached by the network, but that have not been reported experimentally.
Results
We inferred the regulatory network that controls the terminal differentiation of B cells from
experimental data available in the literature referring to the key molecules involved in the con-
trol of terminal B cell differentiation from the precursor B cell (Naive) to GC, Mem or PC cell
types (Fig 2). The network contains 22 nodes representing functional molecules or molecular
complexes, namely AID, Ag, Bach2, Bcl6, BCR, Blimp1, CD40, CD40L, ERK, IL-2, IL-2R, IL-4,
IL-4R, IL-21, IL-21R, Irf4, NF-κB, Pax5, STAT3, STAT5, STAT6 and XBP1. These nodes have
39 regulatory interactions among them, being either positive or negative. S1 Table contains the
set of key references used to infer the regulatory network depicted in Fig 2.
Fig 2. The regulatory network of B cells. Nodes represent molecules or molecular complexes. Positive and
negative regulatory interactions among molecules are represented as green continuous arrows and red blunt
arrows respectively.
doi:10.1371/journal.pcbi.1004696.g002
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
4 / 26
The regulatory network consists of two sets of nodes, i.e., those pertaining to a core module
integrated by the master transcriptional regulators of terminal B cell differentiation (Bach2, Bcl6,
Blimp1, Irf4, Pax5 and XBP1) and a set of nodes representing several signal transduction cas-
cades (Ag/BCR/ERK, CD40L/CD40NF-κB, IL-2/IL-2R/STAT5, IL-4/IL-4R/STAT6 and IL-21/
IL21R/STAT3) representing key external signals required for the control of the differentiation
process. The nodes corresponding to these signaling pathways are active if a external stimuli is
present, i.e., if a extracellular molecule is available, it is recognized by a specific receptor that
transduce the signal by a messenger molecule, which in turn regulates the expression of the tran-
scription factors in the core regulatory network. Most interactions among the nodes in Fig 2 were
inferred from the literature. However, we found necessary to incorporate to the network the
interactions Pax5 ! Bcl6, Irf4 a Pax5 and the self-regulatory interactions Bcl6 ! Bcl6, and Pax5
! Pax5 so as to obtain attractors with biological significance. Therefore, these four regulatory
interactions constitute predictions of our model. The following paragraphs resume the reasons to
incorporate such unreported interactions into our regulatory network model.
B cells develop in the bone marrow from hematopoietic progenitors that progressively lose
its multipotent potential as they commit with the B cell lineage. This process strictly depends
on the expression of Pax5, which induces chromatin changes of B cell specific genes and
restricts the developmental potential of lymphoid progenitors by repressing genes associated
with other cell type programs [30, 31]. Pax5 is upregulated at the onset of B cell development
until differentiation to plasma cells [7]. During early stages of B cell development, Pax5 expres-
sion is positively controlled by the transcription factor Ebf1 [32], which in turn is activated by
Pax5 [33], thus conforming a mutually activatory regulatory circuit that controls B cell identity.
However, the signals that maintain Pax5 expression throughout late stages of B cell differentia-
tion are not well understood. Therefore, a positive autoregulatory interaction for Pax5 was
included in order to account for the direct mechanisms, possibly via the positive regulatory cir-
cuit between Ebf1 and Pax5, or indirect mechanisms, via other signals, that might sustain high
Pax5 expression during late B cell differentiation.
Once B cells have completed their development in the bone marrow, they migrate to the
bloodstream into the secondary lymphoid organs where they complete maturation throughout
the germinal center reaction. The transcription factor Bcl6 is essential for germinal center for-
mation, since Bcl6 deficiency results in the absence of germinal centers in mice [9, 15]. Given
that Pax5 is required from the beginning of B cell development [10], it was necessary to include
a positive regulatory signal from Pax5 to Bcl6 to keep Bcl6 in an active state when the Pax5
node is active.
A high expression of Bcl6 is required during the GC phase where it controls the expression
of genes necessary for the germinal center program, such as DNA damage response and apo-
ptosis, thus promoting the processes of SHM and CSR and cell proliferation [6]. It has been
shown that mutations that disrupt a negative autoregulatory circuit deregulate Bcl6 expression
and contribute to extensive proliferation in dense large B cell lymphoma (DLBCL) [34]. More-
over, it has been reported that in normal conditions there exist epigenetic mechanisms associ-
ated with positive regulation of Bcl6 expression during the GC phase that overcome its
negative autoregulation [35, 36]. However, the precise mechanisms and signals that maintain
high levels of Bcl6 in GCs are not fully understood. Therefore, we found necessary to include a
positive autoregulatory interaction for Bcl6 in order to account for the possible role of these
mechanisms in GC cell differentiation.
Direct contact of B cells with T cells mediated by the union of the CD40 receptor with its
ligand CD40L induce the expression of Irf4 [16]. It has been shown that low levels of Irf4 pro-
mote the early B cell program, while high Irf4 levels inhibit GC program and promote differen-
tiation toward the Mem or PCs in later stages of B cell differentiation [18, 37]. Given that Pax5
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
5 / 26
is an essential regulator of the B cell identity program [10], a negative interaction between Pax5
and Irf4 was incorporated to simulate a constant activation of the Pax5 circuit when the Irf4
node is low and to inhibit Pax5 activation when Irf4 present at high levels.
Attractors of the wild type network
We studied the dynamical behavior of the discrete and the continuous systems so as to obtain
their attractors. The discrete version of the B cell regulatory network was studied by exhaus-
tively testing the behavior of the network from all possible initial conditions. The system
reaches exactly four fixed point attractors, shown in Table 1. Notably, there is a one-to-one
relation of these four attractors with the expression patterns of the cell types shown in Fig 1.
We labeled the attractors as Naive, GC, Mem, and PC. It is important to remember that
each attractor represents a different configuration pattern of the network at the steady state.
Specifically, the first attractor, where the nodes Pax5 and Bach2 are active, can be interpreted
as the activation pattern of Naive cells. The second attractor, with high levels of Bcl6, Pax5, and
Bach2, corresponds to the GC cell type. The third attractor, with high levels of Irf4, Pax5, and
Bach2, along with the absence of Bcl6 can be interpreted as the Mem cell fate. Finally, the
fourth attractor, with high Blimp1, Irf4, and XBP1, corresponds to the pattern of the cell type
PC (Fig 3).
In the discrete version of the model, the set of initial states draining to the attractors, i.e the
basins of attraction, do not partition the state space evenly. The percentage of initial states lead-
ing to each of the attractors were as follows: Naive = 56.25%, GC = 6.25%, Mem = 6.25%,
PC = 31.25%. The size of the basins reflects how an attractor can be attained from different ini-
tial configurations, and may indicate the relative stability of such steady state [38]. It has been
suggested that different basins represent stable or semistable cellular differentiation states [29].
Moreover, in order to transit form one steady state to another, a specific external signal would
need to trigger a response in order to overcome the basin of attraction such that a different
attractor could be reached by the system. Configurations with larger basins can be easily
reached from many initial states, therefore, different perturbations could be buffered and cana-
lized by the network towards a particular steady state [39].
Since the Naive and PC states have larger basins than that for GC or Mem attractors, it is
possible to suggest that the former are relatively more stable than the later. Importantly, the
proportion of basin sizes of the Naive, GC and Mem attractors agree with in vivo measures of B
Table 1. Attractors of the discrete and continuous models of the B cell regulatory network.
Naive
GC
Mem
PC
Node
Disc.
Cont.
Disc.
Cont.
Disc.
Cont.
Disc.
Cont.
Bach2
1
1.0E + 0
1
1.0E + 0
1
1.0E + 0
0
1.1E −22
Bcl6
0
1.8E −22
1
1.0E + 0
0
1.4E −22
0
1.0E −22
Blimp1
0
2.3E −22
0
1.3E −22
0
1.3E −22
1
1.0E + 0
Irf4
0
1.7E −22
0
9.7E −23
1
1.0E + 0
1
1.0E + 0
Pax5
1
1.0E + 0
1
1.0E + 0
1
1.0E + 0
0
1.0E −22
XBP1
0
2.3E −22
0
1.2E −22
0
9.9E −23
1
1.0E + 0
For the continuous system we present averages from a total of 500,000 runs from random initial states. The standard deviation are smaller than 1E−22 in
all cases. For simplicity, only the nodes conforming the network core are shown. The rest of nodes belong to the signal transduction cascades, and all of
them are in the inactive state, i.e. 0.
doi:10.1371/journal.pcbi.1004696.t001
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
6 / 26
cells where the Naive progenitor is more abundant in proportion than the other three cell types
[40, 41]. However in spite of the low abundance of PC cells in vivo as a result of a selection pro-
cess of B lymphocytes during the germinal center reaction, the largest basins corresponding to
the progenitor Naive and terminally differentiated PC cells suggest that the regulatory network
assures the formation of these cell types in a robust manner.
Contrary to discrete systems, continuous dynamical systems have an infinite number of
possible initial states so that the search for attractors by sampling a large number of random
initial states can lead to the possibility to miss attractors with small basins of attraction. Indeed,
the sampling of initial states resulted in the finding of only four attractors for the continuous
model, which resulted identical to the attractors of the discrete model, see Table 1. Therefore,
to find possible missing attractors we made an exhaustive perturbation study by temporarily
modifying the activation state of each node in the four attractors found by random sampling
[42]. With this approach we found three more fixed point attractors in the continuous model.
These extra attractors are characterized by intermediate values of activation of the nodes con-
forming the network core and do not have a counterpart in the discrete model, since the dis-
crete model can attain only 0 or 1 activation values (Table 2).
These attractors with intermediate values may represent possible unstable activation states
that can be reached by the system but have not been yet experimentally observed or may corre-
spond to transient differentiation states. Indeed, one of the attractors (“New3” attractor) found
in Table 2 shows intermediate levels of Bcl6 and Irf4, in spite of the antagonistic role of these
two factors, suggesting that low levels of Irf4 controls the establishment of stationary states
prior to Bcl6 downregulation. This attractor may correspond to the known activation pattern
of centrocytes, which are Irf4int, Bcl6hi B cells exiting the GC reaction that represent an inter-
mediate cellular state between GC and PC cells [43]. This result supports the role of Irf4 as a
regulator of the differentiation process prior the terminal differentiation to PCs since it has
been observed that intermediate levels of Irf4 promote the GC program, whereas high levels of
Irf4 promote Bcl6 downregulation and further PC differentiation as B cells exit the germinal
center [44].
Fig 3. Attractors and cell types The stationary states of the regulatory network correspond to multiple
activation patterns that characterize different cell types.
doi:10.1371/journal.pcbi.1004696.g003
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
7 / 26
The differentiation process
The B cell regulatory network is able to describe the differentiation process outlined in Fig 1,
from the Naive precursor to any of the GC, Mem, or PC cell types by means of sequential
pulses of extracellular signals known to direct terminal B cell differentiation (Fig 4). The system
is initialized starting from the Naive attractor, and the system is perturbed at a time t 25 with
a single high pulse of IL-2 or IL-4 for 2 or more time units. Computationally, this is achieved
by fixing the variable IL −4 = 1 and the equation dIL4
dt
¼ 0 for the indicated period of time.
This signal was intended to mimic the effect of subjecting the Naive cell to a saturating extra-
cellular concentration of IL-2 or IL-4 for a brief incubation time. After the pulse, the entire sys-
tem was left to evolve until it converged. This perturbation is sufficient to move the dynamical
system to the GC attractor which is in agreement with the observations that IL-2 and IL-4 pro-
mote B cell proliferation and germinal center formation, and are also necessary signals for the
transition of Naive B cells to GC B cells [45–47].
Differentiation of either Naive or GC cells to Mem cells is mediated by the activation of the
CD40 receptor by its ligand CD40L [48], which leads to Irf4 induction and to the repression of
Bcl6 [16]. Our model recovers these differentiation routes with a saturating activation of
CD40L for 2 or more abitrary time units, which leads to the activation of Irf4 node when the
Table 2. Fixed point attractors of the continuous system not found in the random search.
Attractor
Node
New1
New2
New3
AID
0
0
0
Ag
0
0
0
Bach2
1
1
1
Bcl6
0.5
0
0.5
BCR
0
0
0
Blimp1
0
0
0
CD40
0
0
0
CD40L
0
0
0
ERK
0
0
0
IL-2
0
0
0
IL-2R
0
0
0
IL-4
0
0
0
IL-4R
0
0
0
IL-21
0
0
0
IL-21R
0
0
0
Irf4
0
0.5
0.5
NF-κB
0
0
0
Pax5
1
1
1
STAT3
0
0
0
STAT5
0
0
0
STAT6
0
0
0
XBP1
0
0
0
Three fixed point attractors were found with the perturbation analysis that has not been found in the
random search. These attractors are characterized by intermediate values of the nodes.
doi:10.1371/journal.pcbi.1004696.t002
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
8 / 26
Pax5 node is active and Blimp1 is not present. Activation of Irf4 downregulates Bcl6 and
directs the transition from the GC to the Mem attractor of the dynamical system, see Fig 4.
Similarly, starting from any of the Naive, GC, or Mem attractors, the system is able to move
to the PC attractor by applying a saturating signal of either IL-21 or Ag. This is consistent with
the experimental reports where BCR activation by Ag induce Blimp1 upregulation, as well as
Pax5 and Bcl6 downregulation thus promoting plasma cell differentiation from either Naive,
GC, or Mem cell types [49–51]. This process is facilitated by the presence of IL-21 which is
transduced by STAT3 [52, 53].
For both the discrete and continuous models we obtained the same biological relevant tran-
sition paths that describe the wild type differentiation pattern outlined in Fig 1. However, given
that the continuous model has 7 fixed-point attractors, its complete fate map is larger than that
for the discrete model (S1 Fig). Nonetheless, the continuous model also presents the known
biologically relevant transitions.
It has been suggested that progression toward a terminal differentiated state involves several
epigenetic changes that reduce the options of a cell to differentiate to other cell types, possibly
by several mechanisms that constraint the function of the components of a regulatory network
thus reducing the dimensionality of the state space and controlling the compartmentalization
of this space into basins of attraction with different sizes [29]. Therefore, the presence of exter-
nal signals could affect the way the nodes of the network activate in response to these signals
which in turn regulate the activation of multiple parts of the network to control the establish-
ment of stationary states of the system and the transitions between these states. Interestingly,
Fig 4. Differentiation from Naive to the PC cell type. The changes in the activation of all nodes of the
network are shown as a heatmap which scales from blue to red as the activation level goes from 0 to 1,
respectively. Extracellular signals are simulated as a burst for two or more units of time (arrows). Starting from
the Naive (Bach2+, Pax5+) stationary state (t = 0 to t 25), the system moves to the GC attractor (Bach2+,
Bcl6+, Pax5+) due to the presence of a simulated pulse of IL-4 (t 25) which in turn transit to the Mem
attractor (Bach2+, Irf4+, Pax5+) due to the action of CD40L (t 55) and finally, Mem attractor moves to the PC
state (Blimp1+, Irf4+) by the presence of Ag signal (t 75).
doi:10.1371/journal.pcbi.1004696.g004
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
9 / 26
no transitions from the PC state to other attractors were obtained in any of the two models,
suggesting that the network controls B cell differentiation towards an effector cell fate in an
irreversible manner while allowing the transition between precursor cell fates (Fig 5).
Simulation of mutants
To gain further insight of the dynamical behavior of the B cell regulatory network we systemat-
ically simulated all possible single loss- and gain-of-function mutants and evaluated the sever-
ity of each mutation by comparing the resulting attractors with those of the wild type model.
Loss-of-function mutations were simulated by fixing at 0 the value of a node, whereas gain-of-
function was simulated by fixing at 1 the same activation state of a node. For each mutant, its
attractors were found, exhaustively in the case of the discrete model, and for the continuous
Fig 5. Complete fate map. Nodes represent the fixed point attractors, and the edges correspond to all the possible single-node perturbations able to move
the system from one attractor to another. For the continuous model, perturbations are simulated by temporarily change the value of a single node to 0, 1 or
0.5, represented by the symbols “−”, “+” and “int”, respectively. For example, IL-2+ means that a temporal activation of IL-2 is able to cause the system to
move from the Naive attractor to the GC attractor. Biologically relevant differentiation routes are represented as blue arrows.
doi:10.1371/journal.pcbi.1004696.g005
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
10 / 26
version, by running the dynamical system from 5000 random initial states and solving the
equations numerically until the system converged. Tables 3, 4 and 5 shows that the mutants
can be grouped according to whether its effect results in the loss of one or more attractors with
respect to the wild type model or if it results in the appearance of atypical attractors not found
in the wild type model.
Importantly, both the discrete and continuous versions of the model were able to describe
most of the reported mutants for the six master regulators that conform the core of the net-
work. For instance, the simulated loss-of-function of the Blimp1 node results in the disappear-
ance of the PC attractor, which is in accordance with the experimentally acknowledged role of
Blimp1 as an essential regulator for PC differentiation [54]. Although absence of Blimp1 in B
cells impedes PC differentiation, it does not affect the establishment of Naive, GC or Mem cell
types [54–56], which is in turn reflected by the model since the network reaches all the Naive,
GC and Mem attractors in spite of the loss-of-function of the Blimp1 node (Table 3).
Additionally, for Blimp1 null mutant a distinct attractor was found showing low Pax5 and
high Irf4 levels. It has been reported that Pax5 inactivation along with Irf4 induction precedes
Blimp1 expression and while Irf4 activation is not sufficient to rescue PC differentiation in the
absence of Blimp1, the coordinate expression of both factors is necessary for complete terminal
Table 3. Simulated null mutant attractors.
Mutant
model
Obtained pattern
Effect
References
Bach2
[0, 0, 0, 0, 1, 0] Naive-like
Only similar attractors to the wild type fates were found.
[9, 12, 19]
[0, 1, 0, 0, 1, 0] GC-like
[0, 0, 0, 1, 1, 0] Mem-like
[0, 0, 1, 1, 0, 1] PC
Bcl6
[1, 0, 0, 0, 1, 0] Naive
Loss of GC attractor.
[13–15, 57, 58]
[1, 0, 0, 1, 1, 0] Mem
[0, 0, 1, 1, 0, 1] PC
Blimp1
[1, 0, 0, 0, 1, 0] Naive
Loss of PC attractor. A distinct attractor with high Irf4 levels found.
[54, 56, 59]
[1, 1, 0, 0, 1, 0] GC
[1, 0, 0, 1, 1, 0] Mem
[0, 0, 0, 1, 0, 0] Other
Irf4
[1, 0, 0, 0, 1, 0] Naive
Only Naive and GC attractors are reached by the network. Loss of Mem and PC
attractors.
[18, 37, 60, 61]
[1, 1, 0, 0, 1, 0] GC
Pax5
[0, 0, 1, 1, 0, 1] PC
Inactivation of Pax5 drives the system to the PC state. An attractor not reported in
literature was found.
[8, 30, 62, 63]
[0, 0, 0, 0, 0, 0] Other
XBP1
[1, 0, 0, 0, 1, 0] Naive
Mild effect over the PC attractor. Naive, GC and Mem attractors are not affected
[64, 65]
[1, 1, 0, 0, 1, 0] GC
[1, 0, 0, 1, 1, 0] Mem
[0, 0, 1, 1, 0, 0] PC-like
Null mutant attractors. The attractors found for each null mutant model and the literature supporting its effect are summarized, for simplicity, only the
patterns of activation for the nodes that conform the core of the network, namely Bach2, Bcl6, Blimp1, Irf4, Pax5 and XBP1 are shown. The steady state
pattern for each mutant is shown in the following order: [Bach2, Bcl6, Blimp1, Irf4, Pax5, XBP1].
doi:10.1371/journal.pcbi.1004696.t003
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
11 / 26
B cell differentiation [56]. Therefore, this attractor may represent a cellular state prior to the
PC state.
Similarly to the Blimp1 null mutation, the simulated gain-of-function mutants for the Pax5,
Bcl6 or Bach2 nodes also result in the loss of the PC attractor but the other three wild type acti-
vation patterns are still reached by the network (Table 4), the constitutive activation of any of
these nodes maintains the system in attractors corresponding to precursor B cell fates, in accor-
dance with the observations showing that forced expression of Pax5 or Bach2 in mature B cells
inhibit terminal differentiation to PCs and are required to maintain the B cell identity program
[12, 66, 67]. Moreover, for the Bach2 gain-of-function model an additional attractor was
found. This attractor is characterized by high levels of Bach2 and Irf4 and low Pax5 in a pattern
similar to the Mem attractor, this attractor may correspond to a state previous to PC differenti-
ation where Bach2 avoids Blimp1 activation when Pax5 is inactive [56, 68].
The simulated Irf4 loss-of-function results in the loss of PC and Mem cell attractors (See
Table 3). Since Irf4 deficient B cells are unable to differentiate into Mem and PCs, the attractors
Table 4. Simulated constitutive mutant attractors.
Mutant
model
Obtained pattern
Effect
References
Bach2
[1, 0, 0, 0, 1, 0]
Naive
Loss of PC attractor. An attractor with active Bach2 and Irf4 was found.
[12]
[1, 1, 0, 0, 1, 0] GC
[1, 0, 0, 1, 1, 0]
Mem
[1, 0, 0, 1, 0, 0]
Other
Bcl6
[1, 1, 0, 0, 1, 0] GC
Only GC and similar attractor are reached.
[34, 66, 69–
71]
[1, 1, 0, 1, 1, 0]
Other
[0, 1, 0, 1, 0, 0]
Other
Blimp1
[0, 0, 1, 1, 0, 1] PC
The system stays in the PC state. Loss of Naive, GC and Mem attractors.
[5, 72–76]
Irf4
[1, 0, 0, 1, 1, 0]
Mem
The system reaches only the Mem and PC attractors.
[18, 44, 70]
[0, 0, 1, 1, 0, 1] PC
Pax5
[1, 0, 0, 0, 1, 0]
Naive
Loss of PC attractor, the other three wild type activation patterns are reached by the network.
[63, 67, 77,
78]
[1, 1, 0, 0, 1, 0] GC
[1, 0, 0, 1, 1, 0]
Mem
XBP1
[1, 0, 0, 0, 1, 1]
Naive-like
Activation of XBP1 node does not affects the establishment of any of the Naive, GC, Mem or PC
attractors. Only similar attractors to the wild type patterns were found.
[54]
[1, 1, 0, 0, 1, 1]
GC-like
[1, 0, 0, 1, 1, 1]
Mem-like
[0, 0, 1, 1, 0, 1] PC
Constitutive mutant attractors. The attractors found for each mutant model and the literature supporting its effect are summarized, for simplicity, only the
patterns of activation for the nodes that conform the core of the network, namely Bach2, Bcl6, Blimp1, Irf4, Pax5 and XBP1 are shown. The steady state
pattern for each mutant is shown in the following order: [Bach2, Bcl6, Blimp1, Irf4, Pax5, XBP1].
doi:10.1371/journal.pcbi.1004696.t004
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
12 / 26
found for this mutant support the role of Irf4 in the formation of PC and Mem cell types [18,
37, 60, 61]. Induction of Irf4 promotes the formation of Mem cells and PC differentiation [18,
44, 70], which is also described by the model as simulated gain-of-function of the Irf4 recovers
only two attractors corresponding to the Mem and PC states. Therefore, constitutive activation
of the Irf4 node drives the system to the Mem and PC cell fate states.
Conversely, constitutive activation of the Bcl6 node results into three attractors, one of them
corresponds to the GC cell pattern, the other two attractors correspond to patterns where Bcl6
is active along with Irf4. These activation patterns coincide with the expression patterns
observed for centrocytes, which are Bcl6+ Irf4+ B cells exiting from the GC reaction [79]. This
result suggest that sustained activation of the Bcl6 node drives the system to a GC or GC-like
state, in accordance with the reported observations where Bcl6 enforced expression in B cells
blocks terminal differentiation and regulates GC formation [34, 66, 71, 80].
Bach2 null mutation does not affects the formation of any of the Naive, GC, Mem or PC cell
types, thus confirming its role as a dispensable regulator of B cell terminal differentiation, but a
necessary negative regulator for Blimp1 expression and PC formation. Only similar attractors
to the wild type fates were found [9, 12, 19].
Bcl6 null mutant mice does not form GC cells but differentiation to Naive, Mem or PC cell
types is not affected. Also, Bcl6-deficient B cells can differentiate into Mem cells or PC inde-
pendently of germinal center reactions. Accordingly the GC attractor is lost in the simulated
Bcl6 loss-of-function mutant [13–15, 57, 58].
Deletion of Pax5 in mice results in the loss of B cells from early pro-B stage. Inactivation of
Pax5 in mature B cells results in the repression of genes necessary for B cell identity. Pax5 defi-
cient B cells differentiate towards the PC cell fate and show Blimp1 up-regulation. Conditional
inactivation of Pax5 in mice mature B cells promotes differentiation toward PCs, in line with
Table 5. Summary of the simulated mutants and external signals.
Mutant models and simulated signals
Resulting attractors with respect to
the wild type model
Bach2+, Bcl6+, Blimp1−, Irf4−, Pax5+
Loss of PC attractor
Bcl6+, IL-2+, IL-2R+, STAT5+, IL-4+, IL-4R+, STAT6+, Irf4+,
CD40L+, CD40+, NF-κB+, Blimp1+, Ag+, BCR+, ERK+, IL-21+, IL-
21R+, STAT3+
Loss of Naive attractor
Bach2+, Bach2−, XBP1+
Replaced Naive, Mem and GC
attractor by similar ones
Bcl6−, IL-21+, IL-21R+, STAT3+, Ag+, BCR+, ERK+, CD40L+,
CD40+, NF-κB+
Loss of GC attractor
Bcl6+, IL-4+, IL-4R+, STAT6+
Only the GC and GC-like attractors are
found
Blimp1−, Pax5−
Atypical attractor found
Blimp1+, Ag+, BCR+, ERK+, IL-21+, IL-21R+, STAT3+
Only PC attractor is found
Irf4+, CD40L+, CD40+, NF-κB+
Only Mem and PC attractors are found
Irf4−
Loss of Mem and PC attractors
Irf4−, Ag+, BCR+, ERK+
Loss of Mem attractor
XBP1+
Replaced PC attractor by a similar one
Effect of all possible single gain- and loss-of-function mutants of the B cell model with respect to wild type,
as reflected by their type of attractors. Symbols “+” and “−” after a node name denote gain-of-function and
loss-of-function mutations, respectively. The effect of the continued activation of the nodes pertaining to
signaling pathways is also indicated with the symbol “+” and summarized in the table.
doi:10.1371/journal.pcbi.1004696.t005
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
13 / 26
the PC attractor found for this mutant. An attractor not reported in literature was found which
may correspond to the total loss of expression of the B cell lineage factors [8, 30, 62, 63].
XBP1 is not strictly required for initiation of PC cell differentiation or for previous differen-
tiation stages of terminal B cell differentiation. The network reaches all the wild type attractors
[64, 65].
Forced expression of Blimp1 promotes terminal differentiation to PC cells. Only the PC
attractor was found for this simulated mutant [5, 72–76].
Loss-of-function of XBP1 affects subsequent PC development but it does not impairs B cell
differentiation or the establishment of any of the Naive, GC, Mem and PC cell types [54].
Accordingly, similar attractors to the wild type patterns were found.
It is important to note that not all single loss- or gain-of-function mutants have a severe
effect on the dynamics of our B cell differentiation model, since simulated Bach2 and XBP1
constitutive and null mutations result in attractors similar to the wild type, suggesting that
these nodes have only a mild effect on the global behavior of the network. However, the Bach2
node is not dispensable since the constitutive activation of this node avoided the network for
reach the PC attractor, in accordance with its biological role as an inhibitor of PC differentia-
tion [12]. These results show the contribution of each node to the dynamics of network and
therefore indicate the importance of these factors as regulators of the differentiation process.
Given that the expression patterns defining each cell type are controlled by the core module
of the regulatory network, the attractors found for the wild-type models as well as for the single
loss- and gain-of-function mutants persist even in the absence of external signals. However, as
mentioned in the above paragraphs, external stimuli can drive the system from one steady state
to another, thus affecting the way the network controls the establishment of different expres-
sion patterns. Therefore, we simulated the continuous presence of external signals by fixing the
activation value of the nodes representing signaling pathways, namely Ag, BCR, CD40, CD40L,
ERK, IL-2, IL-2R, IL-4, IL-4R, IL-21, IL-21R, NF-κB, STAT3, STAT5, and STAT6, in order to
analyze how its continued activation influences the behavior of the core regulatory network
affecting the appearance and maintenance of multiple cell fates. For clarity, the effect of the
continued stimulation by external signals and the effect of the simulated mutants on the sta-
tionary patterns reached by the network is summarized in Table 5.
Discussion
The hematopoietic system is well characterized at the cellular level, and there exist several
efforts to reconstruct and analyze parts of its underlying molecular regulatory network to
understand the differentiation process of multiple cell types. Network modeling has become an
appropriate tool for the systematic study of the dynamical properties of specific regulatory net-
works and signaling pathways. The dynamic behavior of even relatively simple networks is nei-
ther trivial nor intuitive. Moreover, experimental information about the kinetic parameters of
the molecules conforming such networks is generally lacking. However, the use of qualitative
methods shows that it is possible to predict the existence of expression patterns or pointing at
missing regulatory interactions.
The model presented in this work describes the activation states observed experimentally
for Naive, GC, Mem and PC cell types. This model is also able to describe the differentiation
pattern from Naive B cells to GC, Mem and PC subsets in response to specific external signals.
Despite the lack of qualitative information it was possible to reconstruct the regulatory network
of B cells and propose a basic regulatory architecture. This model propose the existence of
some missing regulatory interactions and activation states not documented in the literature
that might play an important role in the context of terminal B cell differentiation. Importantly,
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
14 / 26
these interactions constitute specific predictions that can be tested experimentally. It is also rel-
evant to stress that the proposed regulatory interactions might be attained by way of intermedi-
ary molecules not included in the regulatory network. This is so because the whole network
modeling approach is based upon the net effect of one node over another, focusing on whether
the flow of information is known, rather than relying on the direct physical contact between
molecules. Furthermore, the results suggest that the dynamical behavior of the B cell regulatory
network is to a large extent determined by the structure of the network rather than the detail of
the kinetic parameters, in accordance to analyses of related models [42, 81, 82].
While Boolean networks constitute a valuable modeling approach of choice whenever there is
only qualitative data available, for this biological system we wanted to incorporate qualitative
continue variables that in addition to the identification of the stationary states as in the discrete
model, allows for the analysis of the effect of gradients of external signals. The dynamical behav-
ior of the model resembles the qualitative behavior of the differentiation process by recovering
the transition of the system from a Naive state to the terminally differentiated PC state under the
presence of external signals. This result recapitulates the directional and branched nature of B
cell differentiation events and supports the key role of extracellular signals in the maintenance
and instruction of the differentiation process. Importantly, the model allows the exploration of
system transitions that describe the differentiation form one cell type to another, it is interesting
to note that no transitions from the PC state to other attractors were obtained, suggesting that
the B cell regulatory network assures the differentiation towards an effector cell fate in an irre-
versible manner whereas allowing plasticity of the precursor cell fates.
There are several ways in which our model could be improved in future versions. One gen-
eral change may be the implementation of the model as a stochastic dynamical system.
Although both the stochastic and deterministic models retain the same steady states, the imple-
mentation as a stochastic system could be useful to generate information about the probability
of the cells to transit from one state to another.
Another possible route of refinement of the models would be the inclusion of a specific time
scale. Both the discrete and continuous models presented here use qualitative modeling frame-
works, with results having arbitrary time units. In order to incorporate phenomena with spe-
cific timescales, it will be necessary either to calibrate the continuous dynamical system by
scanning for appropriate values for the parameters, or alternatively make use of a quantitative
modeling framework. Also, it possible to add other layers of regulation to the model, for exam-
ple by incorporating the effect of chromatin remodeling on the availability of some genes.
However, given that we were able to recover with a small qualitative network the basic patterns
of activation, it is possible that the role played by the levels of regulation not included in the
present model may significantly reduce the number of possible transitory trajectories of the
system, instead of determining nature and number of the stationary states themselves.
Finally, despite the qualitative nature of the model presented here, we believe it might be
used as seed to analyze important biological and clinical phenomena, given that deregulation
of the master regulators included in the network are known to be involved in oncogenic events
occurring in multiple lymphomas. For instance, aberrant expression of Bcl6 may lead to consti-
tutive repression of genes necessary for exit of the GC program and normal differentiation,
therefore contributing to lymphomagenesis [83]. In addition, activation of Irf4 leads to exten-
sive cell proliferation and survival [84]. The present model could serve as a starting framework
to test different hypothesis regarding the possible routes by which the expression of the afore-
mentioned factors and other components of the network could be regulated in order to find
therapeutic intervention strategies or to test how deregulation of the known mechanisms could
lead to pathological conditions, thus contributing to our knowledge on the development of
lymphomas.
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
15 / 26
Materials and Methods
Molecular basis of the B cell regulatory network
We inferred the regulatory network controlling terminal B cell differentiation from experimen-
tal data available in literature. The evidence used to recover the nodes and interactions of the B
cell regulatory network (Fig 2) is summarized in the following paragraphs. The transition from
Naive B cells to GC, Mem, and antibody-secreting PCs is regulated by the coordinated activity
of transcription factors that act as key regulators of the differentiation process. These factors
appear to regulate mutually antagonistic genetic programs and can be divided into those that
promote and maintain the B cell program, such as Pax5, Bcl6, and Bach2, and those that con-
trol terminal differentiation into memory cells or plasma cells, such as Irf4 and Blimp1 and
XBP1 [7].
Pax5 functions as the master regulator of B cell identity, it is expressed at the onset of B cell
differentiation and is maintained in all developmental stages of B cells upon commitment to
plasma cells. Pax5-deficiency results in the loss of B cell identity and the acquisition of multili-
neage potential [10]. Pax5 directly inhibits Blimp1 transcription by binding to the promoter of
Prdm1 the gene encoding Blimp1 [11]. In turn, Blimp1 represses Pax5 [78], thus conforming a
mutually exclusive regulatory circuit. Along with Pax5, Bach2 avoids PC differentiation and
promotes class switch recombination by repressing Blimp1 through binding to a regulatory ele-
ment on the Prdm1 gene [12]. Bach2 is positively regulated by Pax5 [31], while being repressed
by Blimp1 in PCs, thus creating a mutual inhibition feedback loop [19].
Bcl6 expression is induced upon arrival of Naive B cells into the germinal centers. Bcl6 is a
transcription factor essential for germinal center formation, since deficiency of Bcl6 results in
the absence of germinal centers in mice [14, 15]. The signals that promote high Bcl6 expression
in GC cells are not fully understood. However, it has been shown that mutations that disrupt a
negative autoregulatory circuit of Bcl6 deregulate its expression and promote the proliferation
of GC cells in dense large B cell lymphomas (DLBCL) [34]. Moreover, it has been reported that
there exists a positive regulatory mechanism controlling high Bcl6 expression during the GC
phase that overcome its negative autoregulation [35, 36]. In accordance with these data, we
found necessary to include in our model a positive autoregulatory interaction for Bcl6 (Bcl6 !
Bcl6) in order to account for the required signals that maintain high Bcl6 activation levels in
GC cells.
Additionally, the presence of IL-2 and IL-4 produced by follicular T helper cells play an
important role in the transition from Naive to GC cells, as these signals are required for the
maintenance and proliferation of GC cells. IL-2/IL-2R and IL-4/IL-4R signals are transduced
by STAT5 and STAT6, respectively, thus positively regulating the expression of Bcl6 [46]. Bcl6
binds directly to the Prdm1 promoter and down-regulates the expression of Blimp1 in GC
cells, thus preventing the terminal differentiation to PCs [85]. Conversely, Bcl6 is a direct target
of Blimp1. This creates a mutual inhibition circuit among Bcl6 and Blimp1 [86]. Maturation of
GC cells towards the Mem or PC cell fates requires the downregulation of Bcl6 [17]. This pro-
cess also depends on the activation of BCR by Ag recognition, as well as on the direct contact
of B cells with T helper cells which leads to BCR activation and the proteosomal degradation of
Bcl6, mediated by ERK [49].
The direct contact between B and T cells is mediated by the union of CD40 with its ligand
CD40L, which in turn activates NF-κB, a positive regulator of Irf4 [16]. Irf4 is a key regulator
required for the development of Mem cells from Naive and GC cells, and is involved in the con-
trol of CSR and PC differentiation [18, 37]. It has been shown that low levels of Irf4 promote
CSR while high Irf4 levels promote PC differentiation. Irf4 inhibits Bcl6 by binding to a regula-
tory site in the Bcl6 gene promoter in response to the direct contact of B and T cells [16].
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
16 / 26
Conversely, Bcl6 is a direct negative regulator of Irf4 in GC cells [87, 88], thus generating a
mutual inhibition circuit between Bcl6 and Irf4. Moreover, high Irf4 expression is maintained
through direct binding of Irf4 to its own promoter creating a positive autoregulatory circuit [61].
Irf4 also plays an important role in early stages of B cell development where it regulates
Pax5 expression through the formation of molecular complexes in the Pax5 enhancer region
[89]. Similarly, Pax5 activation during B cell development is maintained by the transcription
factor Ebf1 [33] which in turn is activated by Pax5 [32], therefore conforming a mutually posi-
tive regulatory circuit. However, the role of the regulatory circuits between Pax5, Irf4 and Ebf1
during terminal B cell differentiation is not clearly understood. Nevertheless, we found neces-
sary to include these interactions in our model (Pax5 ! Pax5 and Irf4 a Pax5) in order to
account for the known activation patterns for these two regulators. Therefore, these interac-
tions constitute predictions of the model that may support an important role of these regula-
tory interactions during the late stages of B cell differentiation.
The processes of CSR and SHM are controlled by the action of AID [90] which is regulated
by the direct binding of Pax5, NF-κB and STAT6 to its regulatory regions in response to IL-4
and CD40 signals [91–93]. AID expression is inhibited in PCs by Blimp1 [5].
Finally, PC differentiation program is regulated by the coordinated activity of Blimp1, Irf4
and XBP1. Blimp1 is specifically expressed in PCs and its activation is sufficient to drive mature
B cell differentiation towards the PC fate [56]. Blimp1 is induced by the direct binding of Irf4
to an intronic region of the Prdm1 gene [18, 61]. Also Blimp1 is involved in Irf4 activation con-
forming a double positive regulatory circuit. Deficient B cells do not express Irf4 and fail to dif-
ferentiate into PCs [94, 95]. In turn, Blimp1 activates XBP1 [64] which is normally repressed
by Pax5 in mature B cells [65].
The regulatory network as a discrete dynamical system
Boolean networks constitute the simplest approach to modeling the dynamics of regulatory
networks. A Boolean network consists of a set of nodes, each of which may attain only one of
two states: 0 if the node is OFF, or 1 if the node is ON [96, 97]. The level of activation for the i-
th node is represented by a discrete variable xi, which is updated at discrete time steps accord-
ing to a Boolean function Fi such that xi(t+1) = Fi[x1(t), x2(t), . . ., xn(t)], where [x1(t), x2(t), . . .,
xn(t)] is the activation state of the regulators of the node xi at time t. The Boolean function Fi is
expressed using the logic operators ^ (AND), _ (OR), and ¬ (NOT). In our model, all Fis are
updated simultaneously, which is known as the synchronous approach. The resulting set of Fis
is shown in Table 6.
We obtained all the attractors of the Boolean model by testing all possible initial states
under a synchronous updating scheme using the R package BoolNet [99]. Moreover, we simu-
lated all possible single loss- and gain-of-function mutants by fixing the value of each node to 0
or 1, respectively.
The complete discrete model is available for testing in The Cell Collective (http://www.
thecellcollective.org/) model B cell differentiation [98]. Furthermore, the model is available as
the accompanying file S1 File (Bcells_model.xml) in SBMLqual format.
The regulatory network as a continuous dynamical system
The B cell regulatory network was converted into a continuous dynamical system by using the
standardized qualitative dynamical systems method (SQUAD) [104, 105] with the modifica-
tion by Sánchez-Corrales and colaborators [42] to include into the equations a version of the
regulatory logic rule for each node. This methodology offers two main advantages, first, it
allows to construct a qualitative model in spite of the lack of kinetic information, making use
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
17 / 26
only of the regulatory interactions of the network, and second, since the external signals are
continuous in nature, this methodology permit to study the response of the network to such
signals while at the same time allowing a direct comparison with the Boolean model. Moreover,
due to its formulation as a set of ordinary differential equations, it may find additional unstable
steady states, cyclic behavior, or attraction basins with respect to Boolean approaches [105].
The SQUAD method approximate a Boolean system with the use of a set of ordinary differ-
ential equations, where the activation level of a node is represented by a variable xi which is
normalized in the range [0, 1]. This is a dimensionless variable since it represents the functional
activation level of a node, but it may be used to represent the normalized concentration of the
active form of a molecule or a macromolecular complex. The change of the xi node over time is
Table 6. Logical rules. The set of Boolean rules defining the regulatory network of the terminal differentiation of B cells.
Logic rule
Description
References
AID (STAT6 _ (NF-κB ^ Pax5)) ^¬
Blimp1
AID node is positively regulated by the presence of Pax5 in response to CD40 and IL-4
signals, transduced by NF-κB and STAT6 respectively. AID is active only if its inhibitor
Blimp1 is absent.
[91–93]
Bach2 Pax5 ^¬ Blimp1
Bach2 node is activated if its positive regulator Pax5 is active and the suppressor Blimp1 is
absent.
[19, 31]
Bcl6 (STAT5 _ STAT6 _ (Pax5
^Bcl6)) ^¬ (Blimp1 _ Irf4 _ ERK)
The node Bcl6 is induced in response to IL-2 and IL-4, transduced by STAT5 and STAT6
respectively. Its activation depends on the presence of Pax5 (proposed as a positive
interaction), and on the mechanisms maintaining its own expression (proposed as a positive
autoregulation). Bcl6 node is repressed if either the nodes Blimp1, Irf4 or ERK are active.
[35, 36, 46]
BCR Ag
BCR node is activated by the input node Ag, simulating the presence of extracellular antigen.
[100]
Blimp1 (ERK _ STAT3) _ (Irf4 ^¬
(Pax5 _ Bcl6 _ Bach2))
Blimp1 is activated by Irf4 if all its inhibitors, Pax5, Bcl6 and Bach2 are inactive. Blimp1 is
induced by Ag and IL-21 which are transduced by ERK and STAT3, respectively.
[49, 50, 52–
54]
CD40 CD40L
The CD40 node is activated by the input node CD40L simulating the direct contact of B with
T cells mediated by the union of the CD40 receptor with its ligand.
[16]
ERK BCR
BCR cross-linking promotes ERK activation after Ag stimulation
[49, 51, 76]
IL-2R IL-2
The IL-2R node is induced by the input node IL-2, simulating the activation of the IL-2R
receptor by IL-2 stimulation, a signal involved in GC differentiation
[4]
IL-4R IL-4
The IL-4 input node induces the IL-4R node simulating the activation of the IL-4R receptor
activation by the cytokine IL-4 required for GC differentiation.
[4]
IL-21R IL-21
The IL-21R receptor is induced by IL-21, a signal required for differentiation toward PCs
[101–103]
Irf4 (NF-κB _ Irf4) _ (Blimp1 ^¬ Bcl6)
Irf4 is induced in response to CD40L signals, transduced by the node NF-κB. Irf4 regulates
its own activation and is positively regulated by Blimp1 if its inhibitor Bcl6 is off.
[16, 18, 61,
89]
NF-κB CD40
Activation of the CD40 receptor promotes the activation of the transcription factor NF-κB in
response to CD40L stimulation
[16]
Pax5 (Pax5 _ ¬ Irf4) ^¬(Blimp1 _
ERK)
Pax5 is maintained active by low levels of Irf4, proposed as a negative interaction, and
possibly by a positive regulatory circuit with Ebf1 that plays a key role during early B cell
differentiation, included as a positive autoregulatory interaction. Pax5 is inhibited if Blimp1 or
ERK are present.
[11, 51, 78]
STAT3 IL-21R
IL-21 signals are transduced by STAT3, represented in the model as a positive interaction of
the IL-21R receptor with STAT3.
[101–103]
STAT5 IL-2R
Activation of the IL-2R receptor by IL-2 induces STAT5 activation
[46]
STAT6 IL-4R
Activation of the IL-4R receptor induces STAT6 in response to IL-4 stimulation
[91]
XBP1 Blimp1 ^¬ Pax5
XBP1 is activated by Blimp1 if the suppressor Pax5 is absent
[65]
The rules determining the state of activation of each node as a function of its regulatory inputs are expressed by the use of the logic operators ^ (AND), _
(OR), and ¬ (NOT).
doi:10.1371/journal.pcbi.1004696.t006
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
18 / 26
controlled by an activation term and a decay term as described by:
dxi
dt ¼
e0:5hi þ ehiðoi0:5Þ
ð1 e0:5hiÞð1 þ ehiðoi0:5ÞÞ gixi
ð1Þ
In Eq (1) parameters hi and γi are the gain of the input of the node and the decaying rate,
respectively. The term ωi is the continuous form of the logical rule describing the response of
the node xi to its regulatory inputs, as defined for the discrete dynamical system in the previous
section. The logical statements defined for the discrete model are converted into their continu-
ous equivalent by changing A ^ B, A _ B, and ¬A in an expression of classic logic into min(A,
B), max(A, B), and 1−A, respectively, thus creating a fuzzy-logic expression. Note that the term
ωi cannot be applied to all nodes of Fig 2, because there are five of them that do not have any
regulatory inputs, therefore equations representing these nodes contain only the term for the
decaying rate.
The activation term for Eq (1) has the form of a sigmoid as a function of the total input to a
node ωi, and was constructed so as to pass through the points (0,0), (0.5,0.5), (1,1) for any posi-
tive value of h. We found that for values of h 50, the curve is very close to a step function; for
intermediate values of h the function is similar to a logistic curve and as h approaches 0 the
function is almost a straight line (Fig 6). This characteristic allows the study of different quali-
tative response curves on the overall behavior of the regulatory network, while at the same time
conserving the direct comparison against a Boolean model due to the three fixed points men-
tioned above. Since there is a lack of published quantitative data that could be used to estimate
the values of either of the hi and γi parameters to solve the system of equations, we decided to
Fig 6. The activation part of Eq (1) is a sigmoid function of the total input of the node (ωi) Regardless
of the value of h, the sigmoid touches the points (0,0), (0.5,0.5) and (1,1). For values of h 50 the curve
resembles a step function.
doi:10.1371/journal.pcbi.1004696.g006
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
19 / 26
use a set of default values. Therefore, all h’s were set to 50 and γ = 1 so as to obtain steep
response curves, thus making an easy comparison of the discrete model against the current
continuous model and/or forthcoming models.
We found that values h 6¼ {4,8} and γ = 1 recover the experimentally observed patterns of
expression S2 Fig. In contrast to the relative insensitivity of changes in the strength of interac-
tions h, the attractors are highly sensitive to changes in values of the decay rate γ. Eq (1) is con-
structed in such a way that γ has to have a value equal to 1 in order for xi’s to lie in the closed
interval [0, 1]. Now, values of γ different than 1 make all attractors to disappear S3 Fig. The
attractors of the B cell regulatory network model, therefore, are highly dependent on the value
used in the parameter specifying the decaying rate.
The resulting dynamical system in shown as S2 Table in the Supporting Information, and
available as the supplementary S1 File. Due to the high non-linearity of the continuous system
of equations, we located the steady states of this model by numerically solving the system of
equations from 500,000 random initial states and letting it converge, with the use of the R pack-
age deSolve [106], the detailed attractors found for both the wild type and the mutant models
are shown in S2 File.
Supporting Information
S1 Table. Table of interactions. Key references supporting network interactions.
(PDF)
S2 Table. The B cell network as a continuous dynamical system. The set of ordinary differen-
tial equations conforming the continuous version of the B cell regulatory network model.
(PDF)
S1 Fig. Complete fate map for the discrete model.
(TIFF)
S2 Fig. Location of the fixed-point attractors as a function of the parameter h.
(TIFF)
S3 Fig. Location of the fixed-point attractors as a function of the parameter γ.
(TIFF)
S1 File. SBMLqual format version of the B cell model. Complete model for testing in the The
Cell Collective platform (http://www.thecellcollective.org/), model B cell differentiation and file
Bcell_model.xml.
(XML)
S2 File. Detailed attractors of wild type and simulated mutants.
(XLSX)
Acknowledgments
We want to thank Carlos Ramírez and Nathan Weinstein for their valuable comments during
the preparation of this manuscript.
Author Contributions
Conceived and designed the experiments: LM. Performed the experiments: AM. Analyzed the
data: AM LM. Contributed reagents/materials/analysis tools: AM. Wrote the paper: AM LM.
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
20 / 26
References
1.
LeBien TW, Tedder TF. B lymphocytes: how they develop and function. Blood. 2008; 112(5):1570–
80. doi: 10.1182/blood-2008-02-078071 PMID: 18725575
2.
Gatto D, Brink R. The germinal center reaction. J Allergy Clin Immunol. 2010; 126(5):898–907; quiz
908–9. doi: 10.1016/j.jaci.2010.09.007 PMID: 21050940
3.
Victora GD, Nussenzweig MC. Germinal centers. Annu Rev Immunol. 2012; 30:429–57. doi: 10.1146/
annurev-immunol-020711-075032 PMID: 22224772
4.
Moens L, Tangye SG. Cytokine-Mediated Regulation of Plasma Cell Generation: IL-21 Takes Center
Stage. Front Immunol. 2014 jan; 5(February):65. doi: 10.3389/fimmu.2014.00065 PMID: 24600453
5.
Shaffer AL, Lin KI, Kuo TC, Yu X, Hurt EM, Rosenwald A, et al. Blimp-1 Orchestrates Plasma Cell Dif-
ferentiation by Extinguishing the Mature B Cell Gene Expression Program. Immunity. 2002 Jul; 17
(1):51–62. doi: 10.1016/S1074-7613(02)00335-7 PMID: 12150891
6.
Basso K, Saito M, Sumazin P, Margolin Aa, Wang K, Lim Wk, et al. Integrated biochemical and
computational approach identifies BCL6 direct target genes controlling multiple pathways in normal
germinal center B cells. Blood. 2010 feb; 115(5):975–84. doi: 10.1182/blood-2009-06-227017 PMID:
19965633
7.
Nutt SL, Taubenheim N, Hasbold J, Corcoran LM, Hodgkin PD. The genetic network controlling
plasma cell differentiation. Semin Immunol. 2011; 23(5):341–9. doi: 10.1016/j.smim.2011.08.010
PMID: 21924923
8.
Delogu A, Schebesta A, Sun Q, Aschenbrenner K, Perlot T, Busslinger M. Gene repression by Pax5
in B cells is essential for blood cell homeostasis and is reversed in plasma cells. Immunity. 2006 Mar;
24(3):269–81. doi: 10.1016/j.immuni.2006.01.012 PMID: 16546096
9.
Muto A, Tashiro S, Nakajima O, Hoshino H, Takahashi S, Sakoda E, et al. The transcriptional pro-
gramme of antibody class switching involves the repressor Bach2. Nature. 2004 Jun; 429(6991):566–
71. doi: 10.1038/nature02596 PMID: 15152264
10.
Cobaleda C, Schebesta A, Delogu A, Busslinger M. Pax5: the guardian of B cell identity and function.
Nat Immunol. 2007; 8(5):463–70. doi: 10.1038/ni1454 PMID: 17440452
11.
Mora-López F, Reales E, Brieva JA, Campos-Caro A, Dc W, Mora-lo F, et al. Human BSAP and
BLIMP1 conform an autoregulatory feedback loop. Blood. 2007; 110(9):3150–7. doi: 10.1182/blood-
2007-05-092262 PMID: 17682124
12.
Ochiai K, Katoh Y, Ikura T, Hoshikawa Y, Noda T, Karasuyama H, et al. Plasmacytic transcription fac-
tor Blimp-1 is repressed by Bach2 in B cells. J Biol Chem. 2006; 281(50):38226–34. doi: 10.1074/jbc.
M607592200 PMID: 17046816
13.
Toyama H, Okada S, Hatano M, Takahashi Y, Takeda N, Ichii H, et al. Memory B Cells without
Somatic Hypermutation Are Generated from Bcl6-Deficient B Cells. Immunity. 2002 Sep; 17(3):329–
339. doi: 10.1016/S1074-7613(02)00387-4 PMID: 12354385
14.
Fukuda T, Yoshida T, Okada S, Hatano M, Miki T, Ishibashi K, et al. Disruption of the Bcl6 gene results
in an impaired germinal center formation. J Exp Med. 1997; 186(3):439–48. doi: 10.1084/jem.186.3.
439 PMID: 9236196
15.
Dent aL, Shaffer AL, Yu X, Allman D, Staudt LM. Control of inflammation, cytokine expression, and
germinal center formation by BCL-6. Science. 1997; 276(5312):589–92. doi: 10.1126/science.276.
5312.589 PMID: 9110977
16.
Saito M, Gao J, Basso K, Kitagawa Y, Smith PM, Bhagat G, et al. A signaling pathway mediating
downregulation of BCL6 in germinal center B cells is blocked by BCL6 gene alterations in B cell lym-
phoma. Cancer Cell. 2007; 12(3):280–92. doi: 10.1016/j.ccr.2007.08.011 PMID: 17785208
17.
Kuo TC, Shaffer AL, Haddad J, Choi YS, Staudt LM, Calame K. Repression of BCL-6 is required for
the formation of human memory B cells in vitro. J Exp Med. 2007; 204(4):819–30. doi: 10.1084/jem.
20062104 PMID: 17403935
18.
Sciammas R, Shaffer aL, Schatz JH, Zhao H, Staudt LM, Singh H. Graded expression of interferon
regulatory factor-4 coordinates isotype switching with plasma cell differentiation. Immunity. 2006; 25
(2):225–36. doi: 10.1016/j.immuni.2006.07.009 PMID: 16919487
19.
Muto A, Ochiai K, Kimura Y, Itoh-Nakadai A, Calame KL, Ikebe D, et al. Bach2 represses plasma cell
gene regulatory network in B cells to promote antibody class switch. EMBO J. 2010; 29(23):4048–61.
doi: 10.1038/emboj.2010.257 PMID: 20953163
20.
Sciammas R, Li Y, Warmflash A, Song Y, Dinner AR, Singh H. An incoherent regulatory network
architecture that orchestrates B cell diversification in response to antigen signaling. Mol Syst Biol.
2011; 7(495):495. doi: 10.1038/msb.2011.25 PMID: 21613984
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
21 / 26
21.
Bhattacharya S, Conolly RB, Kaminski NE, Thomas RS, Andersen ME, Zhang Q. A bistable switch
underlying B-cell differentiation and its disruption by the environmental contaminant 2,3,7,8-tetrachlor-
odibenzo-p-dioxin. Toxicol Sci. 2010; 115(1):51–65. doi: 10.1093/toxsci/kfq035 PMID: 20123757
22.
Zhang Q, Kline DE, Bhattacharya S, Crawford RB, Conolly RB, Thomas RS, et al. All-or-none sup-
pression of B cell terminal differentiation by environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-
dioxin. Toxicol Appl Pharmacol. 2013; 268(1):17–26. doi: 10.1016/j.taap.2013.01.015 PMID:
23357550
23.
Martínez MR, Corradin A, Klein U, Álvarez MJ, Toffolo GM, di Camillo B, et al. Quantitative modeling
of the terminal differentiation of B cells and mechanisms of lymphomagenesis. Proc Natl Acad Sci U S
A. 2012; 109(7):2672–7. doi: 10.1073/pnas.1113019109 PMID: 22308355
24.
Meyer-Hermann M. A mathematical model for the germinal center morphology and affinity maturation.
J Theor Biol. 2002;p. 273–300.
25.
Hawkins JB, Jones MT, Plassmann PE, Thorley-Lawson Da. Chemotaxis in densely populated tissue
determines germinal center anatomy and cell motility: a new paradigm for the development of com-
plex tissues. PLoS One. 2011; 6(12):e27650. doi: 10.1371/journal.pone.0027650 PMID: 22145018
26.
Kestler Ha, Wawra C, Kracher B, Kühl M. Network modeling of signal transduction: Establishing the
global view; 2008.
27.
Morris MK, Saez-Rodriguez J, Sorger PK, Lauffenburger Da. Logic-based models for the analysis of
cell signaling networks; 2010.
28.
Albert R, Thakar J. Boolean modeling: A logic-based dynamic approach for understanding signaling
and regulatory networks and for making useful predictions; 2014.
29.
Huang S, Eichler G, Bar-Yam Y, Ingber DE. Cell fates as high-dimensional attractor states of a com-
plex gene regulatory network. Phys Rev Lett. 2005 Apr; 94(12):1–4. doi: 10.1103/PhysRevLett.94.
128701
30.
Horcher M, Souabni A, Busslinger M. Pax5/BSAP maintains the identity of B cells in late B lymphopoi-
esis. Immunity. 2001 Jun; 14(6):779–90. doi: 10.1016/S1074-7613(01)00153-4 PMID: 11420047
31.
Schebesta A, Mcmanus S, Salvagiotto G, Delogu A, Busslinger GA, Busslinger M. Transcription fac-
tor Pax5 activates the chromatin of key genes involved in B cell signaling, adhesion, migration, and
immune function. Immunity. 2007; 27(1):49–63. doi: 10.1016/j.immuni.2007.05.019 PMID: 17658281
32.
Roessler S, Györy I, Imhof S, Spivakov M, Williams RR, Busslinger M, et al. Distinct promoters medi-
ate the regulation of Ebf1 gene expression by interleukin-7 and Pax5. Mol Cell Biol. 2007 Jan; 27
(2):579–594. doi: 10.1128/MCB.01192-06 PMID: 17101802
33.
O’Riordan M, Grosschedl R. Coordinate regulation of B cell differentiation by the transcription factors
EBF and E2A. Immunity. 1999 Jul; 11(1):21–31. doi: 10.1016/S1074-7613(00)80078-3 PMID:
10435576
34.
Pasqualucci L, Migliazza A, Basso K, Houldsworth J, Chaganti RSK, Dalla-Favera R. Mutations of the
BCL6 proto-oncogene disrupt its negative autoregulation in diffuse large B-cell lymphoma. Blood.
2003; 101(8):2914–23. doi: 10.1182/blood-2002-11-3387 PMID: 12515714
35.
Ramachandrareddy H, Bouska A, Shen Y, Ji M, Rizzino A, Chan WC, et al. BCL6 promoter interacts
with far upstream sequences with greatly enhanced activating histone modifications in germinal cen-
ter B cells. Proc Natl Acad Sci U S A. 2010; 107(26):11930–5. doi: 10.1073/pnas.1004962107 PMID:
20547840
36.
Batlle-López A, Cortiguera MG, Rosa-Garrido M, Blanco R, Del Cerro E, Torrano V, et al. Novel CTCF
binding at a site in exon1A of BCL6 is associated with active histone marks and a transcriptionally
active locus. Oncogene. 2013;(April: ):1–11.
37.
Klein U, Casola S, Cattoretti G, Shen Q, Lia M, Mo T, et al. Transcription factor IRF4 controls plasma
cell differentiation and class-switch recombination. Nature immunology. 2006; 7(7):773–782. doi: 10.
1038/ni1357 PMID: 16767092
38.
Demongeot J, Goles E, Morvan M, Noual M, Sené S. Attraction basins as gauges of robustness
against boundary conditions in biological complex systems. PLoS One. 2010 Jan; 5(8):e11793. doi:
10.1371/journal.pone.0011793 PMID: 20700525
39.
Huang S. The molecular and mathematical basis of Waddington’s epigenetic landscape: A framework
for post-Darwinian biology? BioEssays. 2012 Feb; 34(2):149–157. doi: 10.1002/bies.201100031
PMID: 22102361
40.
Kjeldsen MK, Perez-Andres M, Schmitz A, Johansen P, Boegsted M, Nyegaard M, et al. Multipara-
metric flow cytometry for identification and fluorescence activated cell sorting of five distinct B-cell
subpopulations in normal tonsil tissue. American Journal of Clinical Pathology. 2011; 136(6):960–
969. doi: 10.1309/AJCPDQNP2U5DZHVV PMID: 22095383
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
22 / 26
41.
Perez-Andres M, Paiva B, Nieto WG, Caraux a, Schmitz a, Almeida J, et al. Human peripheral blood
B-Cell compartments: A crossroad in B-cell traffic. Cytometry Part B—Clinical Cytometry. 2010; 78
(SUPPL. 1):47–60. doi: 10.1002/cyto.b.20547
42.
Sánchez-Corrales YE, Alvarez-Buylla ER, Mendoza L. The Arabidopsis thaliana flower organ specifi-
cation gene regulatory network determines a robust differentiation process. J Theor Biol. 2010; 264
(3):971–83. doi: 10.1016/j.jtbi.2010.03.006 PMID: 20303988
43.
Cattoretti G, Shaknovich R, Smith PM, Jäck Hm, Murty VV, Alobeid B. Stages of germinal center tran-
sit are defined by B cell transcription factor coexpression and relative abundance. Journal of immunol-
ogy (Baltimore, Md: 1950). 2006 Nov; 177(10):6930–9. doi: 10.4049/jimmunol.177.10.6930
44.
Ochiai K, Maienschein-Cline M, Simonetti G, Chen J, Rosenthal R, Brink R, et al. Transcriptional Reg-
ulation of Germinal Center B and Plasma Cell Fates by Dynamical Control of IRF4. Immunity. 2013
May; 38(5):918–929. doi: 10.1016/j.immuni.2013.04.009 PMID: 23684984
45.
Holder MJ, Liu YJ, Defrance T, Flores-Romo L, MacLennan IC, Gordon J. Growth factor requirements
for the stimulation of germinal center B cells: evidence for an IL-2-dependent pathway of develop-
ment. Int Immunol. 1991; 3(12):1243–51. doi: 10.1093/intimm/3.12.1243 PMID: 1723294
46.
Scheeren Fa, Naspetti M, Diehl S, Schotte R, Nagasawa M, Wijnands E, et al. STAT5 regulates the
self-renewal capacity and differentiation of human memory B cells and controls Bcl-6 expression. Nat
Immunol. 2005; 6(3):303–13. doi: 10.1038/ni1172 PMID: 15711548
47.
Nojima T, Haniuda K, Moutai T, Matsudaira M, Mizokawa S, Shiratori I, et al. In-vitro derived germinal
centre B cells differentially generate memory B or plasma cells in vivo. Nat Commun. 2011; 2:465. doi:
10.1038/ncomms1475 PMID: 21897376
48.
Arpin C, Déchanet J, Van Kooten C, Merville P, Grouard G, Brière F, et al. Generation of memory B
cells and plasma cells in vitro. Science. 1995; 268(5211):720–2. doi: 10.1126/science.7537388
PMID: 7537388
49.
Niu H, Ye BH, Dalla-Favera R. Antigen receptor signaling induces MAP kinase-mediated phosphory-
lation and degradation of the BCL-6 transcriptionÃ? factor. Genes Dev. 1998; 12(13):1953–1961. doi:
10.1101/gad.12.13.1953 PMID: 9649500
50.
Kwon H, Thierry-Mieg D, Thierry-Mieg J, Kim HP, Oh J, Tunyaplin C, et al. Analysis of interleukin-21-
induced Prdm1 gene regulation reveals functional cooperation of STAT3 and IRF4 transcription fac-
tors. Immunity. 2009; 31(6):941–52. doi: 10.1016/j.immuni.2009.10.008 PMID: 20064451
51.
Yasuda T, Hayakawa F, Kurahashi S, Sugimoto K, Minami Y, Tomita A, et al. B cell receptor-ERK1/2
signal cancels PAX5-dependent repression of BLIMP1 through PAX5 phosphorylation: a mechanism
of antigen-triggering plasma cell differentiation. J Immunol. 2012; 188(12):6127–34. doi: 10.4049/
jimmunol.1103039 PMID: 22593617
52.
Ettinger R, Sims GP, Fairhurst AM, Robbins R, da Silva YS, Spolski R, et al. IL-21 induces differentia-
tion of human naive and memory B cells into antibody-secreting plasma cells. J Immunol. 2005; 175
(12):7867–79. doi: 10.4049/jimmunol.175.12.7867 PMID: 16339522
53.
Berglund LJ, Avery DT, Ma CS, Moens L, Deenick EK, Bustamante J, et al. IL-21 signalling via STAT3
primes human naïve B cells to respond to IL-2 to enhance their differentiation into plasmablasts.
Blood. 2013; 122(24):3940–3950. doi: 10.1182/blood-2013-06-506865 PMID: 24159173
54.
Shapiro-Shelef M, Lin KI, McHeyzer-Williams LJ, Liao J, McHeyzer-Williams MG, Calame K. Blimp-1
Is Required for the Formation of Immunoglobulin Secreting Plasma Cells and Pre-Plasma Memory B
Cells. Immunity. 2003; 19(4):607–620. doi: 10.1016/S1074-7613(03)00267-X PMID: 14563324
55.
Shapiro-Shelef M, Lin KI, Savitsky D, Liao J, Calame K. Blimp-1 is required for maintenance of long-
lived plasma cells in the bone marrow. J Exp Med. 2005; 202(11):1471–6. doi: 10.1084/jem.
20051611 PMID: 16314438
56.
Kallies A, Nutt SL. Terminal differentiation of lymphocytes depends on Blimp-1. Current opinion in
immunology. 2007; 19(2):156–62. doi: 10.1016/j.coi.2007.01.003 PMID: 17291741
57.
Ye BH, Cattoretti G, Shen Q, Zhang J, Hawe N, de Waard R, et al. The BCL-6 proto-oncogene con-
trols germinal-centre formation and Th2-type inflammation. Nat Genet. 1997 Jun; 16(2):161–70. doi:
10.1038/ng0697-161 PMID: 9171827
58.
Huang C, Hatzi K, Melnick A. Lineage-specific functions of Bcl-6 in immunity and inflammation are
mediated by distinct biochemical mechanisms. Nat Immunol. 2013 Mar; 14(4). doi: 10.1038/ni.2543
59.
Shapiro-Shelef M, Lin KI, Savitsky D, Liao J, Calame K. Blimp-1 is required for maintenance of long-
lived plasma cells in the bone marrow. J Exp Med. 2005 Dec; 202(11):1471–6. doi: 10.1084/jem.
20051611 PMID: 16314438
60.
Mittrucker HW. Requirement for the Transcription Factor LSIRF/IRF4 for Mature B and T Lymphocyte
Function. Science. 1997; 275(5299):540–543. doi: 10.1126/science.275.5299.540 PMID: 8999800
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
23 / 26
61.
Shaffer AL, Emre NCT, Lamy L, Ngo VN, Wright G, Xiao W, et al. IRF4 addiction in multiple myeloma.
Nature. 2008; 454(7201):226–31. doi: 10.1038/nature07064 PMID: 18568025
62.
Urbánek P, Wang ZQ, Fetka I, Wagner EF, Busslinger M. Complete block of early B cell differentiation
and altered patterning of the posterior midbrain in mice lacking Pax5/BSAP. Cell. 1994 Dec; 79
(5):901–12. doi: 10.1016/0092-8674(94)90079-5 PMID: 8001127
63.
Nera KP, Kohonen P, Narvi E, Peippo A, Mustonen L, Terho P, et al. Loss of Pax5 promotes plasma
cell differentiation. Immunity. 2006 Mar; 24(3):283–93. doi: 10.1016/j.immuni.2006.02.003 PMID:
16546097
64.
Shaffer AL, Shapiro-Shelef M, Iwakoshi NN, Lee AH, Qian SB, Zhao H, et al. XBP1, downstream of
Blimp-1, expands the secretory apparatus and other organelles, and increases protein synthesis in
plasma cell differentiation. Immunity. 2004; 21(1):81–93. doi: 10.1016/j.immuni.2004.06.010 PMID:
15345222
65.
Todd DJ, McHeyzer-Williams LJ, Kowal C, Lee AH, Volpe BT, Diamond B, et al. XBP1 governs late
events in plasma cell differentiation and is not required for antigen-specific memory B cell develop-
ment. The Journal of experimental medicine. 2009; 206(10):2151–9. doi: 10.1084/jem.20090738
PMID: 19752183
66.
Baron BW, Anastasi J, Montag A, Huo D, Baron RM, Karrison T, et al. The human BCL6 transgene
promotes the development of lymphomas in the mouse. Proc Natl Acad Sci U S A. 2004; 101
(39):14198–203. doi: 10.1073/pnas.0406138101 PMID: 15375218
67.
Usui T, Wakatsuki Y, Matsunaga Y, Kaneko S, Koseki H, Kita T, et al. Overexpression of B cell-spe-
cific activator protein (BSAP/Pax-5) in a late B cell is sufficient to suppress differentiation to an Ig high
producer cell with plasma cell phenotype. J Immunol. 1997; 158(7):3197–204. PMID: 9120274
68.
Kurosaki T, Kometani K, Ise W. Memory B cells. Nature Reviews Immunology. 2015; 15(3):149–159.
doi: 10.1038/nri3802 PMID: 25677494
69.
Cattoretti G, Angelin-Duclos C, Shaknovich R, Zhou H, Wang D, Alobeid B. PRDM1/Blimp-1 is
expressed in human B-lymphocytes committed to the plasma cell lineage. J Pathol. 2005; 206(1):76–
86. doi: 10.1002/path.1752 PMID: 15772984
70.
Teng Y, Takahashi Y, Yamada M, Kurosu T, Koyama T, Miura O, et al. IRF4 negatively regulates pro-
liferation of germinal center B cell-derived Burkitt’s lymphoma cell lines and induces differentiation
toward plasma cells. Eur J Cell Biol. 2007 Oct; 86(10):581–9. doi: 10.1016/j.ejcb.2007.05.006 PMID:
17651861
71.
Reljic R, Wagner SD, Peakman LJ, Fearon DT. Suppression of Signal Transducer and Activator of
Transcription 3-Dependent B Lymphocyte Terminal Differentiation by Bcl-6. J Exp Med. 2000; 192
(12):1841–1848. doi: 10.1084/jem.192.12.1841 PMID: 11120780
72.
Horikawa K, Takatsu K. Interleukin-5 regulates genes involved in B-cell terminal maturation. Immunol-
ogy. 2006 Aug; 118(4):497–508. doi: 10.1111/j.1365-2567.2006.02382.x PMID: 16895557
73.
Turner CA, Mack DH, Davis MM. Pillars article: Blimp-1, a novel zinc finger-containing protein that can
drive the maturation of B lymphocytes into immunoglobulin-secreting cells. 1994. J Immunol. 2010
Jul; 185(1):5–14. doi: 10.4049/jimmunol.1090043 PMID: 20562267
74.
Lin Y. Repression of c-myc Transcription by Blimp-1, an Inducer of Terminal B Cell Differentiation. Sci-
ence. 1997 Apr; 276(5312):596–599. doi: 10.1126/science.276.5312.596 PMID: 9110979
75.
Piskurich JF, Lin KI, Lin Y, Wang Y, Ting JP, Calame K. BLIMP-I mediates extinction of major histo-
compatibility class II transactivator expression in plasma cells. Nat Immunol. 2000 Dec; 1(6):526–32.
doi: 10.1038/82788 PMID: 11101876
76.
Yasuda T, Kometani K, Takahashi N, Imai Y, Aiba Y, Kurosaki T. ERKs induce expression of the tran-
scriptional repressor Blimp-1 and subsequent plasma cell differentiation. Sci Signal. 2011 Jan; 4
(169):ra25. doi: 10.1126/scisignal.2001592 PMID: 21505187
77.
Morrison aM, Nutt SL, Thévenin C, Rolink a, Busslinger M. Loss- and gain-of-function mutations
reveal an important role of BSAP (Pax-5) at the start and end of B cell differentiation. Semin Immunol.
1998 Apr; 10(2):133–42. doi: 10.1006/smim.1998.0115 PMID: 9618759
78.
Lin KI, Angelin-Duclos C, Kuo TC, Calame K. Blimp-1-dependent repression of Pax-5 is required for
differentiation of B cells to immunoglobulin M-secreting plasma cells. Mol Cell Biol. 2002; 22
(13):4771–80. doi: 10.1128/MCB.22.13.4771-4780.2002 PMID: 12052884
79.
Falini B, Fizzotti M, Pucciarini A, Bigerna B, Marafioti T, Gambacorta M, et al. A monoclonal antibody
(MUM1p) detects expression of the MUM1/IRF4 protein in a subset of germinal center B cells, plasma
cells, and activated T cells. Blood. 2000; 95(6):2084–92. PMID: 10706878
80.
Cattoretti G, Pasqualucci L, Ballon G, Tam W, Nandula SV, Shen Q, et al. Deregulated BCL6 expres-
sion recapitulates the pathogenesis of human diffuse large B cell lymphomas in mice. Cancer cell.
2005 May; 7(5):445–55. doi: 10.1016/j.ccr.2005.03.037 PMID: 15894265
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
24 / 26
81.
Martínez-Sosa P, Mendoza L. The regulatory network that controls the differentiation of T lympho-
cytes. Biosystems. 2013; 113(2):96–103. doi: 10.1016/j.biosystems.2013.05.007 PMID: 23743337
82.
Mendoza L, Pardo F. A robust model to describe the differentiation of T-helper cells. Theory Biosci.
2010;p. 283–293.
83.
Shaffer aL, Rosenwald A, Staudt LM. Decision making in the immune system: Lymphoid Malignan-
cies: the dark side of B-cell differentiation. Nat Rev Immunol. 2002 dec; 2(12):920–933. doi: 10.1038/
nri953 PMID: 12461565
84.
Shaffer AL, Young RM, Staudt LM. Pathogenesis of Human B Cell Lymphomas. Annu Rev Immunol.
2012 apr; 30(1):565–610. doi: 10.1146/annurev-immunol-020711-075027 PMID: 22224767
85.
Tunyaplin C, Shaffer aL, Angelin-Duclos CD, Yu X, Staudt LM, Calame KL. Direct repression of
prdm1 by Bcl-6 inhibits plasmacytic differentiation. J Immunol. 2004; 173(2):1158–65. doi: 10.4049/
jimmunol.173.2.1158 PMID: 15240705
86.
Shaffer AL, Yu X, He Y, Boldrick J, Chan EP, Staudt LM. BCL-6 Represses Genes that Function in
Lymphocyte Differentiation, Inflammation, and Cell Cycle Control. Immunity. 2000; 13(2):199–212.
doi: 10.1016/S1074-7613(00)00020-0 PMID: 10981963
87.
Gupta S, Jiang M, Anthony A, Pernis AB. Lineage-specific modulation of interleukin 4 signaling by
interferon regulatory factor 4. The Journal of experimental medicine. 1999; 190(12):1837–1848. doi:
10.1084/jem.190.12.1837 PMID: 10601358
88.
Alinikula J, Nera KP, Junttila S, Lassila O. Alternate pathways for Bcl6-mediated regulation of B cell to
plasma cell differentiation. European journal of immunology. 2011; 41(8):2404–13. doi: 10.1002/eji.
201141553 PMID: 21674482
89.
Decker T, Pasca di Magliano M, McManus S, Sun Q, Bonifer C, Tagoh H, et al. Stepwise Activation of
Enhancer and Promoter Regions of the B Cell Commitment Gene Pax5 in Early Lymphopoiesis.
Immunity. 2009 Apr; 30(4):508–520. doi: 10.1016/j.immuni.2009.01.012 PMID: 19345119
90.
Muramatsu M, Kinoshita K, Fagarasan S, Yamada S, Shinkai Y, Honjo T. Class switch recombination
and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing
enzyme. Cell. 2000; 102(5):553–63. doi: 10.1016/S0092-8674(00)00078-7 PMID: 11007474
91.
Dedeoglu F. Induction of activation-induced cytidine deaminase gene expression by IL-4 and CD40
ligation is dependent on STAT6 and NF B. International Immunology. 2004; 16(3):395–404. doi: 10.
1093/intimm/dxh042 PMID: 14978013
92.
Gonda H, Sugai M, Nambu Y, Katakai T, Agata Y, Mori KJ, et al. The balance between Pax5 and Id2
activities is the key to AID gene expression. The Journal of experimental medicine. 2003; 198
(9):1427–37. doi: 10.1084/jem.20030802 PMID: 14581609
93.
Tran TH, Nakata M, Suzuki K, Begum NA, Shinkura R, Fagarasan S, et al. B cell-specific and stimula-
tion-responsive enhancers derepress Aicda by overcoming the effects of silencers. Nature immunol-
ogy. 2010; 11(2):148–54. doi: 10.1038/ni.1829 PMID: 19966806
94.
Kallies A, Hasbold J, Tarlinton DM, Dietrich W, Corcoran LM, Hodgkin PD, et al. Plasma cell ontogeny
defined by quantitative changes in blimp-1 expression. The Journal of experimental medicine. 2004;
200(8):967–77. doi: 10.1084/jem.20040973 PMID: 15492122
95.
Sciammas R, Davis MM. Modular nature of Blimp-1 in the regulation of gene expression during B cell
maturation. The Journal of Immunology. 2004; 172(9):5427–40. doi: 10.4049/jimmunol.172.9.5427
PMID: 15100284
96.
Kauffman SA. Metabolic stability and epigenesis in randomly constructed genetic nets. J Theor Biol.
1969 Mar; 22(3):437–467. doi: 10.1016/0022-5193(69)90015-0 PMID: 5803332
97.
Wang RS, Saadatpour A, Albert R. Boolean modeling in systems biology: an overview of methodology
and applications; 2012.
98.
Helikar T, Kowal B, McClenathan S, Bruckner M, Rowley T, Madrahimov A, et al. The Cell Collective:
Toward an open and collaborative approach to systems biology. BMC systems biology. 2012; 6(1):96.
doi: 10.1186/1752-0509-6-96 PMID: 22871178
99.
Müssel C, Hopfensitz M, Kestler Ha. BoolNet—an R package for generation, reconstruction, and anal-
ysis of Boolean networks. Bioinformatics. 2010; 26(10):1378–1380. doi: 10.1093/bioinformatics/
btq124 PMID: 20378558
100.
Schmidt C, Kim D, Ippolito GC, Naqvi HR, Probst L, Mathur S, et al. Signalling of the BCR is regulated
by a lipid rafts-localised transcription factor, Bright. EMBO J. 2009 Mar; 28(6):711–24. doi: 10.1038/
emboj.2009.20 PMID: 19214191
101.
Zotos D, Coquet JM, Zhang Y, Light A, D’Costa K, Kallies A, et al. IL-21 regulates germinal center B
cell differentiation and proliferation through a B cell-intrinsic mechanism. J Exp Med. 2010 Feb; 207
(2):365–78. doi: 10.1084/jem.20091777 PMID: 20142430
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
25 / 26
102.
Linterman MA, Beaton L, Yu D, Ramiscal RR, Srivastava M, Hogan JJ, et al. IL-21 acts directly on B
cells to regulate Bcl-6 expression and germinal center responses. J Exp Med. 2010 Feb; 207(2):353–
63. doi: 10.1084/jem.20091738 PMID: 20142429
103.
Rankin AL, MacLeod H, Keegan S, Andreyeva T, Lowe L, Bloom L, et al. IL-21 Receptor Is Critical for
the Development of Memory B Cell Responses. J Immunol. 2011 Jan; 186(2):667–74. doi: 10.4049/
jimmunol.0903207 PMID: 21169545
104.
Mendoza L, Xenarios I. A method for the generation of standardized qualitative dynamical systems of
regulatory networks. Theor Biol Med Model. 2006; 3:13. doi: 10.1186/1742-4682-3-13 PMID:
16542429
105.
Weinstein N, Mendoza L. Building Qualitative Models of Plant Regulatory Networks with SQUAD.
Front Plant Sci. 2012; 3(April):72. doi: 10.3389/fpls.2012.00072 PMID: 22639661
106.
Soetaert K, Petzoldt T, Setzer R. Solving Differential Equations in R. R J. 2010; 2(December):5–15.
Network Model of B Cells
PLOS Computational Biology | DOI:10.1371/journal.pcbi.1004696
January 11, 2016
26 / 26
|
26751566
|
XBP1 = ( Blimp1 AND ( ( ( NOT Pax5 ) ) ) )
Blimp1 = ( ( ( ( Irf4 AND ( ( ( NOT Bcl6 OR NOT Bach2 OR NOT Pax5 ) ) ) ) AND NOT ( Bach2 ) ) AND NOT ( Pax5 ) ) AND NOT ( Bcl6 ) ) OR ( ( ( ( ERK ) AND NOT ( Bach2 ) ) AND NOT ( Pax5 ) ) AND NOT ( Bcl6 ) ) OR ( ( ( ( STAT3 ) AND NOT ( Bach2 ) ) AND NOT ( Pax5 ) ) AND NOT ( Bcl6 ) )
Bach2 = ( ( Pax5 ) AND NOT ( Blimp1 ) )
IL-21R = ( IL-21 )
STAT3 = ( IL-21R )
CD40 = ( CD40L )
Bcl6 = ( ( ( ( Bcl6 AND ( ( ( Pax5 ) ) ) ) AND NOT ( Blimp1 ) ) AND NOT ( ERK ) ) AND NOT ( Irf4 ) ) OR ( ( ( ( STAT5 ) AND NOT ( Blimp1 ) ) AND NOT ( ERK ) ) AND NOT ( Irf4 ) ) OR ( ( ( ( STAT6 ) AND NOT ( Blimp1 ) ) AND NOT ( ERK ) ) AND NOT ( Irf4 ) )
STAT5 = ( IL-2R )
Irf4 = ( ( Blimp1 ) AND NOT ( Bcl6 ) ) OR ( ( Irf4 ) AND NOT ( Bcl6 ) ) OR ( NF-kB )
AID = ( STAT6 AND ( ( ( NOT Blimp1 ) ) ) ) OR ( NF-kB AND ( ( ( Pax5 ) ) AND ( ( NOT Blimp1 ) ) ) )
IL-2R = ( IL-2 )
IL-4R = ( IL-4 )
Pax5 = ( ( ( ( Pax5 AND ( ( ( NOT Irf4 ) ) OR ( ( Pax5 ) ) ) ) AND NOT ( ERK ) ) AND NOT ( Blimp1 ) ) ) OR NOT ( Irf4 OR Pax5 OR ERK OR Blimp1 )
NF-kB = ( CD40 )
STAT6 = ( IL-4R )
ERK = ( BCR )
BCR = ( Ag )
|
ORIGINAL RESEARCH
published: 14 April 2016
doi: 10.3389/fgene.2016.00044
Frontiers in Genetics | www.frontiersin.org
1
April 2016 | Volume 7 | Article 44
Edited by:
Ekaterina Shelest,
Hans-Knöll-Institute, Germany
Reviewed by:
Julio Vera González,
University Hospital Erlangen, Germany
Nils Blüthgen,
Charité-Universitätsmedizin
Berlin, Germany
*Correspondence:
Hauke Busch
h.busch@dkfz.de;
Melanie Boerries
m.boerries@dkfz.de
†Present Address:
Steffen Knauer,
Cold Spring Harbor Laboratory, Cold
Spring Harbor, NY, USA
‡These authors are co-first authors.
§These authors are co-last authors.
Specialty section:
This article was submitted to
Bioinformatics and Computational
Biology,
a section of the journal
Frontiers in Genetics
Received: 20 November 2015
Accepted: 14 March 2016
Published: 14 April 2016
Citation:
Offermann B, Knauer S, Singh A,
Fernández-Cachón ML, Klose M,
Kowar S, Busch H and Boerries M
(2016) Boolean Modeling Reveals the
Necessity of Transcriptional Regulation
for Bistability in PC12 Cell
Differentiation. Front. Genet. 7:44.
doi: 10.3389/fgene.2016.00044
Boolean Modeling Reveals the
Necessity of Transcriptional
Regulation for Bistability in PC12 Cell
Differentiation
Barbara Offermann 1‡, Steffen Knauer 1 †‡, Amit Singh ‡, María L. Fernández-Cachón 1,
Martin Klose 1, Silke Kowar 1, Hauke Busch 1, 2, 3*§ and Melanie Boerries 1, 2, 3*§
1 Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research,
Albert-Ludwigs-University Freiburg, Freiburg, Germany, 2 German Cancer Consortium, Freiburg, Germany, 3 German Cancer
Research Center, Heidelberg, Germany
The nerve growth factor NGF has been shown to cause cell fate decisions toward either
differentiation or proliferation depending on the relative activity of downstream pERK,
pAKT, or pJNK signaling. However, how these protein signals are translated into and
fed back from transcriptional activity to complete cellular differentiation over a time span
of hours to days is still an open question. Comparing the time-resolved transcriptome
response of NGF- or EGF-stimulated PC12 cells over 24 h in combination with protein
and phenotype data we inferred a dynamic Boolean model capturing the temporal
sequence of protein signaling, transcriptional response and subsequent autocrine
feedback. Network topology was optimized by fitting the model to time-resolved
transcriptome data under MEK, PI3K, or JNK inhibition. The integrated model confirmed
the parallel use of MAPK/ERK, PI3K/AKT, and JNK/JUN for PC12 cell differentiation.
Redundancy of cell signaling is demonstrated from the inhibition of the different MAPK
pathways. As suggested in silico and confirmed in vitro, differentiation was substantially
suppressed under JNK inhibition, yet delayed only under MEK/ERK inhibition. Most
importantly, we found that positive transcriptional feedback induces bistability in the cell
fate switch. De novo gene expression was necessary to activate autocrine feedback that
caused Urokinase-Type Plasminogen Activator (uPA) Receptor signaling to perpetuate
the MAPK activity, finally resulting in the expression of late, differentiation related genes.
Thus, the cellular decision toward differentiation depends on the establishment of a
transcriptome-induced positive feedback between protein signaling and gene expression
thereby constituting a robust control between proliferation and differentiation.
Keywords: PC12 cells, Boolean modeling, NGF signaling, EGF signaling, bistability
1. INTRODUCTION
The rat pheochromocytoma cells PC12 are a long established in vitro model to study neuronal
differentiation, proliferation and survival (Greene and Tischler, 1976; Burstein et al., 1982; Cowley
et al., 1994). After stimulation with the nerve growth factor (NGF), a small, secreted protein
from the neurotrophin family, PC12 cells differentiate into sympathetic neuron-like cells, which is
Offermann et al.
Boolean Model of PC12 Cell Differentiation
morphologically marked by neurite outgrowth over a time course
of up to 6 days (Levi-Montalcini, 1987; Chao, 1992; Fiore et al.,
2009; Weber et al., 2013). NGF binds with high affinity to the
TrkA receptor (tyrosine kinase receptor A), thereby activating
several
downstream
protein
signaling
pathways including
primarily the protein kinase C/phospholipase C (PKC/PLC),
the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT)
and the mitogen-activated protein kinase/extracellular signal-
regulated kinase (MAPK/ERK) pathways (Kaplan et al., 1991;
Jing et al., 1992; Vaudry et al., 2002). Beyond these immediate
downstream pathways, further studies showed the involvement
of Interleukin 6 (IL6), Urokinase plasminogen activator (uPA)
and Tumor Necrosis Factor Receptor Superfamily Member 12A
(TNFRSF12A) in PC12 cell differentiation (Marshall, 1995; Wu
and Bradshaw, 1996; Leppä et al., 1998; Xing et al., 1998; Farias-
Eisner et al., 2000, 2001; Vaudry et al., 2002; Tanabe et al., 2003).
Sustained ERK activation is seen as necessary and sufficient for
the successful PC12 cell differentiation under NGF stimulation
(Avraham and Yarden, 2011; Chen et al., 2012), whereas transient
ERK activation upon epidermal growth factor (EGF) stimulation
results in proliferation (Gotoh et al., 1990; Qui and Green, 1992;
Marshall, 1995; Vaudry et al., 2002). In fact, selective pathway
inhibition or other external stimuli that modulate the duration of
ERK activation likewise determine the cellular decision between
proliferation and differentiation (Dikic et al., 1994; Vaudry et al.,
2002; Santos et al., 2007). Consequently, the MAPK signaling
network, as the key pathway in the cellular response, has been
studied thoroughly in vitro and in silico (Sasagawa et al., 2005;
von Kriegsheim et al., 2009; Saito et al., 2013). Interestingly,
both EGF and NGF provoke a similar transcriptional program
within the first hour. Therefore, differences in cellular signaling
must be due (i) to differential regulation of multiple downstream
pathways and (ii) late gene response programs (>1 h) that
feed back into the protein signaling cascade. As an example for
pathway crosstalk, both, the MAPK/ERK and c-Jun N-terminal
kinase (JNK) pathways regulate c-Jun activity and are necessary
for PC12 cell differentiation (Leppä et al., 1998; Waetzig and
Herdegen, 2003; Marek et al., 2004), while uPA receptor (uPAR)
signaling, as a result of transcriptional AP1 (Activator Protein-1)
regulation, is necessary for differentiation of unprimed PC12 cells
(Farias-Eisner et al., 2000; Mullenbrock et al., 2011).
In
the
present
study,
we
combined
time-resolved
transcriptome analysis of EGF and NGF stimulated PC12
cells up to 24 h with inhibition of MAPK/ERK, JNK/JUN, and
PI3K/AKT signaling, to develop a Boolean Model of PC12 cell
differentiation that combines protein signaling, gene regulation
and autocrine feedback. The Boolean approach allows to derive
important predictions without detailed quantitative kinetic
data and parameters over different time scales (Singh et al.,
2012). Protein signaling comprised MAPK/ERK, JNK/JUN,
and PI3K/AKT pathways. Based on the upstream transcription
factor analysis and transcriptional regulation of Mmp10 (Matrix
Metallopeptidase 10), Serpine1 (Serpin Peptidase Inhibitor,
Clade E, Member) and Itga1 (Integrin, Alpha 1), we further
included an autocrine feedback via uPAR signaling. The model
topology was trained on the transcriptional response after
pathway inhibition. Inhibition of JNK completely blocked
PC12 cell differentiation and long-term expression of target
transcription factors (TFs), such as various Kruppel-like factors
(Klf2, 4, 6 and 10), Maff(V-Maf Avian Musculoaponeurotic
Fibrosarcoma Oncogene Homolog F) and AP1. Interestingly,
inhibition of MEK (mitogen-activated protein kinase kinase),
blocking the phosphorylation of ERK, slowed down, but not
completely abolished cell differentiation. Neurite quantification
over 6 days confirmed a late and reduced, but significant PC12
differentiation, which hinted at alternative pathway usage
through JNK. Inhibition of the PI3K/AKT pathway, which is
involved in cell proliferation (Chen et al., 2012), even increased
the neuronal morphology and neurite outgrowth.
In conclusion, our Boolean modeling approach shows the
complex interplay of protein signaling, transcription factor
activity and gene regulatory feedback in the decision and
perpetuation of PC12 cell differentiation after NGF stimulation.
2. MATERIALS AND METHODS
2.1. Cell Culture and Stimulation
PC12 cells were obtained from ATCC (American Type Culture
Collection, UK) and were cultured at 37◦C at 5% CO2 in
RPMI 1640 medium, supplemented with 10% Horse Serum, 5%
Fetal Bovine Serum, 1% penicillin/streptomycin (PAN Biotech,
Germany) and 1% glutamine (PAN Biotech, Germany). For
cell stimulation, 500,000 cells/well were seeded on collagen
coated 6 well plates (Corning, NY, USA). The following day,
cells were stimulated with 50 ng/ml rat nerve growth factor
(NGF; Promega, Madison, WI, USA) or 75 ng/ml epidermal
growth factor (EGF; R&D Systems; Wiesbaden, Germany)
for the corresponding times. For the pathway inhibition
experiments, the following inhibitors were used and added 60
min before NGF was added, mitogen-activated protein inhibitor
at a concentration of 20 µM (MEKi; U0126 from Promega,
Madison, WI, USA), phosphoinositide 3-kinase inhibitor at a
concentration of 40 µM (PI3Ki; LY-294002 from Enzo Life
Sciences, New York, USA) and c-Jun N-terminal kinase inhibitor
at a concentration of 20 µM (JNKi; SP600125 from Sigma-
Aldrich, St. Louis, USA). The inhibitors were dissolved in DMSO
and were further diluted in cell culture medium at their working
concentration. Control cells were treated with DMSO at the
same concentration that was present in the cells with inhibitor
treatment.
2.2. RNA Isolation and Quantitative Real
Time PCR (qRT-PCR)
Total RNA was isolated from 500,000 cells per timepoint
according to the manufacturer’s protocol (Universal RNA
Purification
Kit,
Roboklon,
Germany).
RNA
integrity
was measured using an Agilent Bioanalyzer-2000 (Agilent
Technologies GmbH, Waldbronn, Germany), and its content
quantified by NanoDrop ND-1000 (Thermo Fisher Scientific,
Wilmington, USA). For RT-qPCR, double strand cDNA was
synthesized from 1 µg of total RNA using the iScriptTM cDNA
Synthesis kit (Quanta Biosciences, Gaithersburg, USA) according
to the manufacturer instructions. RT-qPCR was performed in
a CFX96 instrument (BioRad, Hercules, CA, USA) using a
Frontiers in Genetics | www.frontiersin.org
2
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
SYBR Green master mix. Relative gene expression levels were
calculated with the 2-11Ct method, using HPRT1 and 18S
ribosomal RNA as reference genes. Post-run analyses were
performed using Bio-Rad CFX Manager version 2.0 and the
threshold cycles (Cts) were calculated from a baseline subtracted
curve fit. See Supplementary Table 1 for primer pair sequences.
2.3. Microscopy and Quantification
Live phase contrast images from PC12 cells under the different
conditions were acquired using a Nikon Eclipse Ti Inverted
Microscope (Nikon; Düsseldorf, Germany) equipped with a
Perfect Focus System (PFS) and a Digital cooled Sight Camera
(DS-QiMc; Nikon, Germany) as described in (Weber et al., 2013).
Briefly, PC12 cells were cultured in collagen coated 6-well plates
(500,000 cells/well) and treated as described in “Cell culture and
stimulation” and 150 images per well, every second day were
recorded with the same spatial pattern. Cell differentiation is
calculated by the ratio of the two described imaging features
(Weber et al., 2013) convex hull (CH) to cell area (CA) for 150
images per well over 6 days (Weber et al., unpublished data).
2.4. Western Blot
For each timepoint and condition 3 × 106 PC12 cells (for
inhibition experiments) or 5 × 106 PC12 cells (for EGF vs.
NGF comparison) were seeded in 10cm collagen coated Cell
BIND dishes (Corning; Germany). Cells were collected after 5,
10, 30 min, 1, 2, 4, 6, 8, 12, 24, and 48 h in 200 µl RIPA
buffer (containing 0.5% SDS), supplemented with proteinase
inhibitor (complete mini EDTA free tablets, Roche, Basel,
Switzerland) and Benzonase (Merck), and lysed for 20 min
under agitation. A total of 30 µg protein was loaded per
lane and run in 10% SDS- polyacrylamide gels, transferred
to polyvinylidene difluoride membranes. Membranes were cut
horizontally into fragments according to the expected sizes
of the protein of interest and immunoblotted with antibodies
against total p44/42 (ERK1/2, 1:2000, #9102S, Cell Signaling
Technology [CST]), phospho p44/42 (pERK1/2, 1:2000, #9101S,
CST), total JNK (JNK1/2, 1:1000, #9258S, CST), phospho JNK
(Thr183/Tyr185, 1:1000, #4668S, CST), total AKT (1:1000,
#4691S, CST), phospho AKT (1:1000, Ser473, #9271S, CST)
or GAPDH (1:2000,# MAB374, Millipore) overnight at 4◦C.
Proteins were visualized with chemiluminescence on SuperSignal
West Pico Chemiluminiscent Substrate imager (Thermo-Fischer,
Massachusetts, USA) after 1h of incubation with appropriate
horseradish
peroxidase-linked
secondary
antibody
(Sigma-
Aldrich). Immunoblots were quantified using ImageJ (image
analyzer camera LAS4000, Fujifilm, Tokyo, Japan). Blots were
normalized to total GAPDH and an internal standard (IS) was
used for normalization between membranes.
2.5. Microarray Analysis and Data
Pre-processing
Time-resolved
gene
expression
data
of
stimulated
PC12
were recorded at t
=
[1, 2, 3, 4, 5, 6, 8, 12, 24] h and
t = [1, 2, 3, 4, 6, 8, 12, 24] h for NGF and EGF stimulation,
respectively.
Control
timepoints
were
measured
at
0, 2, 4, 6, 8, 12, 24 h. Total RNA was isolated, labeled and
hybridized to an Illumina RatRef-12 BeadChip (Illumina, San
Diego, CA, USA) according to the manufacturers protocol.
Raw microarray data were processed and quantile normalized
using the Bioconductor R package beadarray (Ritchie et al.,
2011). Illumina Probes were mapped to reannotated Entrez
IDs using the Illumina Ratv1 annotation data (v. 1.26) from
Bioconductor. If several probes mapped to the same Entrez ID,
the one having the largest interquartile range was retained. This
resulted in 15,348 annotated genes, whose expression was further
batch corrected according to their chip identity (Johnson et al.,
2007). Finally, gene expression time series were smoothed by
a 5th order polynomial to take advantage of the high sampling
rate and replicates at 0, 12, and 24 h. Microarray data have
been deposited at Gene Expression Omnibus (GEO) under the
accession number GSE74327.
2.6. Multi-Dimensional Scaling
To determine significantly regulated genes over time we
performed a multi-dimensional scaling (MDS) using the HiT-
MDS algorithm (Strickert et al., 2005). The algorithm projects
the 15348 × 15348 distance matrix D of the pairwise Euclidean
distances between all genes onto a two dimensional space, while
preserving distances in D as best as possible. Genes varying
strongly and uniquely over time will appear as outliers in the
MDS point distribution. The uniqueness of a gene expression
profile was quantified by fitting a two-dimensional skewed
Gaussian distribution (Azzalini, 2015) to the MDS point density
function.
2.7. Clustering Gene Expression Patterns
To cluster the gene timeseries, we applied the Cluster Affinity
Search Technique (CAST), which considers the genes and their
similarity over times as nodes and weighted edges of graph,
respectively (Ben-Dor et al., 1999). All clusters are considered
as unrelated entities and there is no pre-defined number of
clusters. Instead a threshold parameter, here t = 0.8, determines
the affinity between genes and this the final number of gene
clusters. Inverse or anti-correlative behavior of genes after NGF
or EGF stimulation was determined by fitting a linear model
to the smoothed gene expression. Genes having a significant
slope with opposite sign and an r2
>
0.7 were taken as
anti-correlated.
2.8. Enrichment Analysis of Transcription
Factor Target Gene Sets
Upstream analysis for putative transcription factors regulating
the EGF and NGF transcriptome responses over time were
assessed by a Gene Set Enrichment analysis (Luo et al., 2009)
using paired control to treatment samples for each timepoint
with an overall cutoffq-value < 0.01. As gene sets we used the
transcription factor target lists from the Molecular Signatures
Database (MSigDB, version 5.0) (Subramanian et al., 2005), for
which we mapped the human genes to the rat orthologs using
BiomaRt (Huang et al., 2014).
Frontiers in Genetics | www.frontiersin.org
3
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
2.9. Boolean Model
We used a Boolean model framework for dynamic analysis of
PC12 cell differentiation. Based on our microarray data and
literature knowledge we constructed a highly connected prior
knowledge network (PKN) consisting of 63 nodes and 109
edges (cf. Supplementary Table 2). The R/Bioconductor package
CellNetOptimizer (CNO) (Saez-Rodriguez et al., 2009) was used
to optimize the PKN by reducing redundant nodes, unobservable
states and edges. For this we rescaled the qRT-PCR fold change
values between 0 and 1 and then transformed with a Hill function
f (x) =
xn
xn + kn as suggested in Saez-Rodriguez et al. (2009), where
n = 2 and k = 0.5 denote the Hill coefficient and the threshold,
above which a node is considered “on,” respectively. Changing
the Hill coefficient between 1 ≤n ≤6 did not change the results
qualitatively. Model topology optimization was performed via
the CellNORdt, which allows fitting with time course data. (See
Supplementary Table 3 for stimulus, inhibition and time course
data). We set the maximal CPU run time for the underlying
genetic algorithm (GA) to 100 s and the relative tolerance to
0.01, using default parameters from the CNO otherwise. A
representative evolution of the average and best residual error in
a GA run is depicted in Supplementary Image 1A. The solutions
quickly converge to a quasi steady state within the time window
of simulation of 100 s. The following edges were fixed to prior to
optimization based on literature knowledge: NGF →PI3K, NGF
→RAS, NGF →PLC, AP1 →NPY, MEK/ERK & JNK →Jund,
MEK/ERK & JNK →Junb, Fosl1 & Jund →AP1, Mmp10 →
RAS, RAS →MEK, PLC →MEK. Model optimization was
performed 100 times and edges were retained, if they appeared
in 70% of the runs. This cutoffwas chosen to generate a sparse
network with robust edges. Performing more runs did not change
the results qualitatively (cf. Supplementary Image 1B). Model
simulations were performed using the R/Bioconductor package
BoolNet (Müssel et al., 2010). The reference publications from
which the interactions have been inferred as well as their Boolean
transition functions are listed in Supplementary Table 4.
3. RESULTS
3.1. Gene Response of PC12 Cells Diverges
for NGF and EGF on Long Time Scales
To elucidate the dynamic gene response of NGF and EGF, we
measured the transcriptome dynamics using Illumina RatRef-
12 Expression BeadChips. PC12 cells were either stimulated
with NGF or EGF, and collected at the following timepoints:
1, 2, 3, 4, 5, 6, 8, 12, and 24 h. The unstimulated control samples
(ctrl) were collected in parallel. Gene expression time series were
smoothed by a 5th order polynomial to take advantage of the high
sampling rate. Finally, we mapped array probes to their respective
Entrez IDs, resulting in 15,348 annotated genes.
A bi plot of the principal component analysis (PCA) for
the 1000 most varying genes depicted a clear separation of the
control, NGF and EGF samples. The PCA scores, representing
the NGF and EGF treated samples, showed a qualitatively similar
behavior up to 4 h after stimulation, yet differed markedly beyond
that time (Figure 1A, left). The absolute length and direction of
the PCA loadings (Figure 1A, right) indicate the contribution of
individual genes to the position of the scores. Correspondingly,
several immediate early genes, such as Junb (Jun B Proto-
Oncogene), Fos (FBJ Murine Osteosarcoma Viral Oncogene
Homolog), Ier2 (Immediate Early Response 2), and Egr1 (Early
Growth Response 1) contributed to the early gene response
under both EGF and NGF stimulation, while members of the
uPAR/Integrin signaling complex, such as Mmp13/10/3 (Matrix
Metallopeptidase 13/10/3), Plat (Plasminogen Activator, Tissue)
and Serpine1 (Serpin Peptidase Inhibitor, Clade E, Member 1)
determined, among others, the separation of the NGF from
the EGF trajectory. Loadings that point toward the control and
late EGF response samples, like Cdca7 (Cell Division Cycle
Associated 7) and G0s2 (G0/G1 Switch 2), are clearly related to
cell cycle progression and additionally highlight the difference
in proliferation vs. differentiation. In conclusion, the NGF gene
response, and thus PC12 cell differentiation, must be determined
by late transcriptional feedback events, that trigger and sustain
MAPK/ERK signaling.
Next, we sought to functionally analyze the transcriptional
differences in early and late gene regulation after EGF and
NGF stimulation. For this we selected genes that are (i)
strongly regulated (log2 fold change of < −1.7 or > 1.7 in
two consecutive timepoints) and (ii) have a unique temporal
expression profile according to a multi-dimensional scaling
(MDS) analysis (p-value < 0.01) (cf. Supplementary Image 2).
We found 152 and 402 genes, meeting both criteria, in the
EGF and NGF data, respectively, among which 126 genes are
shared by both conditions. Figure 1B depicts a clustering of these
differentially i.e., top-regulated genes. A cluster affinity search
technique (Ben-Dor et al., 1999) identified five EGF (E1-E3b) and
seven NGF (N1-N4B) gene response clusters (cf. Supplementary
Table 5 and Supplementary Image 3). Interestingly, the EGF
stimulus induced a short pulse-like response with rapid return
to original gene expression levels, while the NGF stimulus
induced a combination of short-impulse like (N1 - N2b) and long
sustained gene expression patterns with several clusters (N3a-
N4b) sustaining their expression over time (cf. circled insets in
Figure 1B).
Figure 1C depicts a network representation of the enrichment
analysis using a hypergeometric test on Gene Ontologies (GO).
Enriched upregulated biological functions were identified in gene
lists E1, E2a, N1, N2a, N3a, N4a and in both groups of inversely
regulated genes (cf. Supplementary Table 6). Nodes correspond to
GO terms, with numbers indicating the joint enrichment scores.
Nodes sharing at least 20 percent of their genes are connected by
solid or dotted edges, if the connected nodes lie within a stimulus
or across NGF and EGF treatment. Early transcription factor
activity is common to both, NGF and EGF signaling, (clusters
E1 and N1) as well as MAPK signaling genes (clusters E2a and
N3a). The latter, however, is more prominent and enriched at
later points in time after NGF stimulation (N3a) compared to the
EGF induced response (E2a). Here, a less and earlier enrichment
of MAPK signaling genes was seen. Moreover, a second network
of transcription factor activity could be identified after NGF
stimulation (cluster N2a) that does not have any equivalent after
EGF stimulation. It seems, that the initial response (first hour) is
Frontiers in Genetics | www.frontiersin.org
4
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
FIGURE 1 | Gene response dynamics after NGF or EGF stimulation. (A) Principal component analysis (PCA) of the PC12 cell transcriptomes after NGF (red),
EGF (blue) and control treatment (gray). The PCA scores (left panel) and loadings (right panel) correspond to the samples and genes, respectively. Samples in the left
panel have been connected to guide the eye. Clearly, EGF and NGF samples remain close in the first 3 h and separate at later timepoints, indicating a different cellular
phenotype. Right panel: 50 largest loading vectors indicating the impact and time of action of individual genes. Immediate early genes, like Fos or Ier2 point toward
early timepoints, while loadings pointing toward the right, like Vgf or Npy, correspond to late timepoints and are most likely involved in differentiation. (B) Expression
clusters of top regulated genes. The left and right panels depict the response of individual genes to EGF and NGF stimulation, respectively (gray lines). Cluster
centroids are marked by lines with the cluster size encoded by line thickness. The circular inserts depict the cluster centroid envelopes for EGF and NGF, respectively.
(C) Network representation of functional enrichment of NGF and EGF response genes. The network is comprised of GO-term clusters having a significant enrichment
(−log10(p-value) > 1.3) as shown in bold black numbers. Red, gray and green nodes contain in this order top-regulated genes, inversely-regulated genes between
EGF and NGF or both. The vertical node location corresponds to the peak regulation of their genes, while node size is proportional to the number of genes in a
functional category. Edges indicate a gene overlap of > 20% between nodes, being drawn as dashed lines, if they are shared between EGF and NGF.
controlled by a shared set of top-regulated genes (cf. Figure 1C,
dashed lines). The cell-fate specific processes, however, seem
to be orchestrated by different set of genes (cf. Figure 1C,
separate networks). Many of the genes executing proliferation
or differentiation specific processes fall into the category of
inversely regulated genes and are not amongst the set of top-
regulated genes identified earlier (cf. Figure 1C, green and gray
nodes, cf. Material and Methods, cf. Supplementary Table 7).
The genes involved in the procession of extracellular matrix
and cytoplasmic vesicles, however, constitute an exception: these
genes are both top and inverse-regulated (cf. Figure 1C, green
nodes).
In summary, functional analysis of the gene clusters revealed
an initiation of the differentiation and proliferation process by
a shared set of differentially regulated genes. Specific functions,
such as transmission of nerve impulse or DNA replication,
however, seemed to be executed by two distinct gene groups
that are when comparing the EGF to the NGF stimulus inversely
regulated over time. Additionally, a second network of genes
involved in transcription factor activity was identified in the
NGF data set, which lacked a corresponding network in the EGF
data set.
3.2. Simulation of a Boolean Network
Based on the above gene response analyses we sought to identify
the mechanisms that sustain MAPK signaling activity after NGF
stimulation. Our transcriptome timeseries analysis revealed that
the decision process between proliferation and differentiation
was spread out over several hours during which transcriptional
feedback through an additional set of transcription factors
was present after NGF stimulation, only (cf. Figure 1C). To
further elucidate the transcription factors upstream of the gene
Frontiers in Genetics | www.frontiersin.org
5
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
response after EGF or NGF stimulation we performed a gene set
enrichment analysis (GSEA) (Luo et al., 2009) on the paired NGF
to control and EGF to control transcriptome timeseries. As gene
sets we used the motif gene sets from the Molecular Signatures
Database (MSigDB v5.0) (Subramanian et al., 2005) and mapped
the human genes onto the rat orthologs using BiomaRt (Huang
et al., 2014).
Figure 2A
compares
the
temporal
significance
of
transcription factors for EGF and NGF stimulation. EGF
elicited an early, yet transient significance of all transcription
factors, while the time-resolved transcription factor significances
for NGF showed early, transient and late activity. Figure 2B
depicts the differences in TF significance between NGF and
EGF. The most down-regulated TFs relative to EGF are E2F1,
EBF1, SOX9 and SP1, all of which are linked to cell proliferation
(Bastide et al., 2007; Hallstrom et al., 2008; Györy et al., 2012;
Zhang et al., 2014).
Mullenbrock et al. (2011) showed late NGF-induced genes up
to 4 h were preferentially regulated by AP1 and CREB (cAMP
response element-binding protein). While AP1 was among the
most persistently up-regulated transcription factors, we found
a transient significance for CREB1, only, peaking at 3 and 6 h,
under EGF or NGF stimulation, respectively, which indicated the
importance of further TFs beyond that time window. In fact,
we found the highest positive differences in the transcription
factors BACH2, AP1, as well as ELF2 and ETV4. The latter
two belong to the ETS transcription factor family. In particular
ETV4, a member of the PEA3 subfamily of ETS, has been
shown to promote neurite outgrowth (Fontanet et al., 2013;
Kandemir et al., 2014). BACH2, member of the BTB-basic region
leucine zipper transcription factor family, is known to down-
regulate proliferation and is involved in neuronal differentiation
of neoblastoma cells via p21 expression (Shim et al., 2006)
and it interacts with the transcription factor MAFF (V-Maf
Avian Musculoaponeurotic Fibrosarcoma Oncogene Homolog
F) (Kannan et al., 2012) that is necessary for differentiation.
To analyze the early cellular response upon treatment, we
additionally compared the phosphorylation levels of pERK,
pAKT and pJNK under NGF and EGF stimulation over time
(Figure 2C). As expected, pERK increased after NGF and EGF
stimulation, showing a persistent up-regulation for 8 h or
pulse-like response, respectively. pJNK was continuously up-
regulated under NGF relative to EGF stimulation, whereas pAKT
responded similar to both stimuli, yet showed a consistently
higher phosphorylation under EGF beyond 2 h. Taken together,
this corroborates the roles of both pERK and pJNK as well as
pAKT in PC12 cell differentiation and proliferation, respectively
(Waetzig and Herdegen, 2003; Chen et al., 2012).
Based on the combined transcriptome, upstream transcription
factor and protein analyses we next developed a comprehensive
prior knowledge interaction network (PKN) for NGF induced
PC12 cell differentiation. The PKN comprises key players of
known pathways involved in PC12 cell differentiation, such
as ERK/PLC/PI3K/JNK/P38/uPAR/NPY and integrin signaling,
as well as “linker nodes” to obtain a minimal, yet fully
connected network, consisting of 63 nodes and 109 reactions
(cf. Supplementary Table 4 for reference publications). The
network is depicted in Supplementary Image 4 with differentially
regulated genes obtained from our timeseries marked in red
and points of inhibition indicated by orange. A Cytoscape
readable network format is provided in Supplementary Table 2.
Albeit the included PKN pathways are much more complex,
our focus was on simulating a biologically plausible signaling
flow, including protein signaling, gene response and autocrine
signaling as follows: stimulated TrkA receptor activates the
downstream pathways PLC/PKC, MAPK/ERK, PI3K/AKT, and
JNK/P38. Phosphorylated ERK, PI3K and P38/JNK together
activate different transcription factors such as Fosl1, Fos, Junb,
Btg2, Klf2/5/6/10, Cited2, Maff, and Egr1, which are important for
PC12 cell differentiation according to our analysis and literature
(Cao et al., 1990; Ito et al., 1990; Levkovitz and Baraban, 2002; Gil
et al., 2004; Eriksson et al., 2007).
Junb and Fos initiate the AP1 system, which in turn
induces uPA/uPAR signaling, triggering the formation of plasmin
(Avraham and Yarden, 2011). The latter is a major factor for
the induction of Mmp3/Mmp10, linking degradation of the
extracellular matrix (ECM) with integrin signaling. The integrins
transmit extracellular signaling back via the focal adhesion kinase
(FAK) (Singh et al., 2012). FAK activates again the SHC protein,
which closes the autocrine signaling. Previous studies reported
that uPAR expression is necessary for NGF-induced PC12 cell
differentiation (Farias-Eisner et al., 2000; Mullenbrock et al.,
2011). A second autocrine signaling loop in the initial PKN
putatively acts via the AP1 system, which in turn activates the
Neuropeptide Y (NPY/NPYY1 pathway). NPY is a sympathetic
co-transmitter that acts via G protein-coupled receptors through
interactions with its NPYY1 receptors (Selbie and Hill, 1998;
Pons et al., 2008). NPYY1 receptor further activates Ca2+
dependent PKC /PLCgamma and subsequently convergences to
ERK signaling.
To
optimize
the
highly
connected
PKN
we
used
CellNetOptimizer (CNO) (Saez-Rodriguez et al., 2009). The
CNO first compresses the network, i.e., it deletes unobservable
nodes and then optimizes the network topology using a genetic
algorithm. We trained the PKN using gene expression of
selected differentially regulated genes under NGF stimulus and
inhibition of either MEK, JNK, or PI3K (Figure 3A, MEKi,
JNKi and PI3Ki). The overall gene response showed a gradual
decline in fold change from NGF via MEK to JNK inhibition,
while inhibition of PI3K only moderately impacted the gene
expression (Figure 3A). The most affected genes under MEK and
JNK inhibition were members of the uPAR signaling pathway,
Mmp10, Mmp3, and Plaur as well as the transcription factors
Fosl1 and Egr1, Plaur, Dusp6 (Dual Specificity Phosphatase 6)
and lastly Npy.
Topology optimization using the above perturbations led to a
greatly reduced network. Optimization lumped linear pathways
into one node, such as the autocrine feedback via uPA/PLAT to
Itga1 and FAK or MEK to ERK transition. The reduced network
revealed both MAPK/ERK and JNK as the central network
hubs, distributing the upstream signals to downstream genes. It
includes two positive feedback via AP1 and uPAR signaling back
to FAK and MAPK as well as AP1 to Npy and PKC/PLC back to
MAPK. To comply with prior knowledge, we re-expanded linear
Frontiers in Genetics | www.frontiersin.org
6
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
FIGURE 2 | Upstream analysis of gene expression timeseries. (A) Upstream Gene Set Enrichment Analysis for transcription factors. The heatmaps depict the
significance of transcription factors putatively controlling the gene response after EGF (left) or NGF stimulation (right). All TFs are significantly regulated (FDR corrected
p-value < 0.01) after NGF treatment. TFs have been clustered by their Euclidean distance across all conditions using a complete linkage method. (B) Difference in TF
p-value significance (NGF-EGF). Rows were ordered from the most positive to the most negative difference at t = 12 and 24 h. (C) Time-resolved quantification of
pERK, pAKT and pJNK after EGF and NGF treatment. Original western blots from PC12 cells treated with 75 ng/ml EGF and 50 ng/ml NGF over time. GAPDH is
shown as loading control, IS: Internal Standard. Statistical analysis of the pERK/ERK, pAKT/AKT and pJNK/JNK levels are shown on the right panel. An increased and
significant higher pERK/ERK level is shown in NGF stimulated (shown as black bars) cells compared to EGF (shown as white bars). A similar trend is visible for
pJNK/JNK. A * denotes a p-value < 0.05, data points obtained in duplicates and triplicates.
Frontiers in Genetics | www.frontiersin.org
7
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
FIGURE 3 | Selective inhibition of NGF-induced PC12 differentiation. (A) Fold change values of selected response genes in PC12 cells after NGF stimulation
under additional inhibition of MEK (NGF+MEKi), JNK (NGF+JNKi), or PI3K (NGF+PI3Ki). Fold change values have been calculated from biological triplicates relative to
the unstimulated control per timepoint. To retain the contrast of less variable genes the maximal fold change has been restrained to +6. Genes have been clustered by
their Euclidean distance across all conditions using a complete linkage method. (B) Optimized Boolean Network based on the training data in (A). Nodes in red have
been measured on the transcript level. Orange nodes indicate inhibited proteins.
pathways and added known down-stream target genes, such
that the final network, shown in Figure 3B, comprised 32 nodes
and 52 edges. We assumed that PC12 differentiation occurs,
if the majority of these genes is activated together with uPAR
signaling. Due to the inherent difficulty of Boolean networks
to incorporate negative feedback loops, we revised the network
topology of the reduced network to include transient gene activity
of several moderately responding genes. Klf4 and Btg2 have been
previously been indicated as immediate early genes in PC12
cell differentiation (Dijkmans et al., 2009) and are involved
in growth arrest (Tirone, 2001; Yoon et al., 2003), which is
a necessary prerequisite for differentiation and degradation of
Frontiers in Genetics | www.frontiersin.org
8
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
mRNA, respectively. While the explicit mechanism of how Klf4
and Btg2 are regulated remains unclear, we assumed an auto-
inhibition once they mediated their growth arrest effect. Zfp36
belongs to the TTP (Tristetraprolin) family of proteins and has
been shown to degrade AU-rich mRNAs, particularly of early
response genes (Amit et al., 2007). It negatively regulates its
own expression (Tiedje et al., 2012) and therefore in the model
effectively delays the activity of AP1 before switching itself off. Of
note, another member of the TTP protein family, Zfp36l2 (zinc
finger protein 36, C3H type-like 2) is constitutively expressed at
long times after NGF stimulation (data not shown) and might
act as another long-term negative feedback regulator and causing
downregulation of Egr1, Fos, and Junb. Indeed, our experimental
data revealed a reduction on gene expression of Egr1, Fos and
Junb over time (Figure 3A).
We simulated the optimized and re-expanded Boolean
network (cf. Supplementary Table 8) using the BoolNet
R/Bioconductor package (Müssel et al., 2010), performing
two types of simulations. First, we tested the robustness and
alternative attractors by setting NGF to “on” and randomly
initializing all other network nodes. The nodes were then
synchronously updated until a steady state was reached. Within
n = 107 different simulations, the same final network state with
“cell differentiation” set to “on” was always reached. Although
this was not an exhaustive search given the number of possible
initial network states, it still demonstrated the robustness of
the network output. Next, to show the information flow from
the NGF receptor to the downstream nodes under different
inhibitory conditions, we initialized all nodes except NGF to
“off” and performed synchronous updates until a steady state
was reached (Figure 4A). Without inhibition, NGF sequentially
switches on MAPK, AKT and JNK pathways as well as uPAR
signaling. Klf4, Btg2, and Zfp36 become transiently active, with
the latter delaying AP1 activity. Blocking MEK (NGF+MEKi)
inhibited ERK and thus several downstream targets, including
the uPAR feedback. As the latter is assumed indispensable for
PC12 cell differentiation, (Farias-Eisner et al., 2000, 2001), the
model predicted inhibition of PC12 cell differentiation. The
same phenotype is found, when blocking JNK (NGF+JNKi). In
comparison to NGF+MEKi it even abrogated the activity of
downstream targets altogether. Inhibition of PI3K (NGF+PI3Ki)
solely affected PI3K and its downstream target protein AKT and
target genes Maffand Klf10, yet cell differentiation persisted.
Taken together, we developed a core network from the
downstream interactome of PC12 cell pathways involved in
differentiation. The model captured the dynamic pathway
activation after NGF stimulation and various inhibitions. It
assigned central and synergistic roles for ERK and JNK in PC12
differentiation with JNK having the largest impact on the network
activity.
3.3. Model Analysis and Experimental
Confirmation
Network simulations were confirmed by live phase-contrast
imaging (Figure 4B) and western blot analyses (Figure 5). We
measured the convex hull (CH) to cell area (CA) ratio of PC12
cells on days 2, 4, and 6. A large convex hull due to extended
neurite (marked as red arrow heads in Figure 4B) and small
overall cell area is indicative of differentiation (Figure 4B, right
panel). Clearly, the continuous CH/CA ratio at day 2 was largest
for NGF stimulation and NGF stimulation with additional PI3K
inhibition, which corresponded well with the cell differentiation
set to “on” in the network simulations under these condition.
One can speculate whether inhibition of the pro-proliferative
PI3K pathway amplifies cell differentiation, possibly relieving
a negative feedback. Indeed, a Western blot of the pERK/ERK
ratio depicted a trend to higher ERK phosphorylation relative
to NGF stimulation under PI3K inhibition (Figure 5) and phase-
contrast images of PC12 cells show more and longer neurites in
comparison to cells treated only with NGF or in combination
to MEKi and JNKi (Figure 4B, NGF+PI3Ki). Interestingly,
image analysis suggested not a stop, but rather a delay of cell
differentiation under MEK inhibition. In detail, PC12 cells show
no neurites under MEKi after 2 days of combined NGF treatment
compared to NGF alone or NGF-PI3Ki. After 4 and 6 days of
NGF+MEKi treatment, less cells have neurites in comparison to
cells that were only treated with NGF (Figure 4B, NGF+MEKi).
In line with literature, pERK levels were reduced, yet pJNK
levels were likewise increased, indicating a redirection of protein
activity under MEK inhibition (Figure 5, right panel). Likewise,
the gene expression showed a reduced, but not completely
abolished fold change for Mmp10 (Figure 3A) and also an up-
regulation of Dusp6. Although the discrete Boolean model could
not simulate gradual responses, MEK inhibition still resulted
in the activation of several downstream target genes necessary
for PC12 cell differentiation, while none of these were active
under JNK inhibition. In summary, modeling and simulation
suggested that PC12 differentiation involved the activity of
both JNK/JUN, MAPK/ERK and PI3K/AKT signaling pathways.
The establishment of a positive, autocrine feedback loop was
indispensable to active late and persistent gene expression.
4. DISCUSSION
PC12 cells are a well established model to study the cellular
decisions toward proliferation or differentiation. Nevertheless,
there is still a lack of understanding on how protein signaling
and gene regulation interact on different time scales to decide
on a long-term, sustained phenotype. Given the fact that PC12
cell cycle and differentiation last up to 4 and 6 days, respectively
(Greene and Tischler, 1976; Luo et al., 1999; Adamski et al., 2007),
late events occurring beyond the first hours are most likely to
be important for sustaining the cellular decision. However, few
studies that have compared the long-term effect of EGF and NGF
in PC12 cells. They focused either on NGF alone (Dijkmans et al.,
2008, 2009), on individual (Angelastro et al., 2000; Marek et al.,
2004; Lee et al., 2005; Chung et al., 2010), or early time-points
(Mullenbrock et al., 2011).
Previous studies have identified expression of immediate
early genes (IEG), such as Egr1, Junb, and Fos together with
delayed early genes (DEG), like Dusp6, Mmp3/10, Fosl1, and
Atf3 as necessary for PC12 cell differentiation (Vician et al.,
Frontiers in Genetics | www.frontiersin.org
9
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
FIGURE 4 | Network simulation of time sequential pathway activation and experimental validation. (A) The heatmaps depict the path to attractor upon NGF
stimulation. Columns correspond to synchronous update steps of the Boolean network. Time progresses from left to right until a steady state is reached. Initially all
nodes, except NGF, are set to zero. Colored boxes correspond to activated nodes with the color denoting individual pathways/node categories. Cells are predicted to
differentiate, if the node “Cell differentiation” is active, as in the case for NGF, or NGF+PI3Ki treatment. (B) Left: phase contrast images for days 2, 4, and 6 are shown
for the 4 different conditions: NGF (control), NGF+MEKi, NGF+PI3Ki and NGF+JNKi. Red arrows depict sites of neurite outgrowth in differentiating PC12 cells. Bar:
100 µm. Right: statistical analysis of PC12 cell differentiation from phase contrast imaging for the different conditions are shown as convex hull (CH) to cell area (CA)
ratio. Bars show Mean ± SEM, n = 2, (*t-test p-value < 0.05).
1997; Levkovitz et al., 2001; Dijkmans et al., 2008; Mullenbrock
et al., 2011). However, we found all these genes strongly
regulated by both EGF and NGF stimulation (Supplementary
Table 5), however, showing differences in their expression
kinetics (Figure 1). Akin to differences in the pERK dynamics,
these results suggest that cellular decisions toward differentiation
or proliferation are driven by the differences in the gene
expression kinetics.
It has been suggested before that distinct cellular stimuli
activate similar sets of response genes, whose expression
dynamics, rather than their composition, determine cellular
decisions (Murphy and Blenis, 2006; Amit et al., 2007; Yosef
and Regev, 2011). Single expression bursts are likely to stimulate
proliferation, while complex, wave-like expression patterns
induce differentiation (Bar-Joseph et al., 2012). Accordingly, EGF
elicited a pulse-like gene response, while NGF induced a complex,
wave-like gene response (Figure 1B). After EGF stimulation the
expression of IEGs, Egr1, Fos, and Junb was quickly attenuated
through the rapid up-regulation of their negative regulators,
namely Fosl1, Atf3, Maff, Klf2, and Zfp36l2 and contributing to
Frontiers in Genetics | www.frontiersin.org
10
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
FIGURE 5 | Quantification of pERK, pAKT and pJNK levels under NGF and individual inhibitor treatments. Determination of pERK/ERK, pAKT/AKT and
pJNK/JNK under NGF, NGF+MEKi, NGF+JNKi and NGF+PI3Ki treatment. Left panel: pERK/ERK levels decrease over time under NGF plus MEK and JNK inhibition.
In contrast, PI3K inhibition shows a similar increase and sustained pERK/ERK levels over time compared to NGF treated PC12 cells alone. Interestingly, pAKT/AKT is
increased under NGF+MEKi treatment, which is particularly significant in the early timepoints (30 min and 1 h) compared to NGF alone or the other two inhibitors. The
latter two show decreased pAKT/AKT levels over time (middle panel). A * denotes a p-value < 0.05, data points obtained in duplicates and triplicates.
a pulse-like gene expression. Furthermore, Fosl1 counteracts Fos
and AP1 (Hoffmann et al., 2005) and Atf3 has been shown to
modulate Egr1 activity (Giraldo et al., 2012), while Maffand
Klf2 negatively regulate serum response and STAT-responsive
promoter elements (Amit et al., 2007). The same genes respond
after NGF stimulation, however with a delayed response and
might be one of the reasons for the stronger and longer gene
and pERK response under NGF stimulation (Murphy et al., 2002,
2004; Murphy and Blenis, 2006; Saito et al., 2013).
A recent study by Mullenbrock et al. (2011) compared
the transcriptome response of PC12 cells to EGF and NGF
stimulation up to 4 h. Using chromatin immunoprecipitation
they found a preferential regulation of late genes through AP1
and CREB TFs after NGF stimulation, which is in line with
our findings (Figure 2A). However, we predicted a constitutive
significance for AP1 up to 24 h, while CREB1 displayed
a transient importance, being most abundant at 6 h after
stimulation. Furthermore, we found a switch in the composition
of transcriptional master regulators between 4 and 12 h. During
this time, late TFs, such as BACH2, ETS1 and ELF2 become
active.
Supplementary Image 5 depicts a Volcano plot of their
target genes. Beyond the early gene targets, such as Fosl1 or
Junb, the late TFs additionally target related to cytoskeleton,
morphogenesis and apoptosis, such as Tumor Necrosis Factor
Receptor Superfamily, Member 12A (Tnfrsf12a), Doublecortin-
Like Kinase 1 (Dclk1), Nerve Growth Factor Inducible Vgf,
Coronin, Actin Binding Protein, 1A (Coro1a, Growth Arrest
And DNA-Damage-Inducible, Alpha (Gadd45a) and Npy. Of
note, we found Rasa2 among the targets, which has recently
been identified as a driver for differentiation through a negative
feedback between PI3K and RAS (Chen et al., 2012).
A recent study by Aoki et al. (2013) investigated the down-
stream gene response upon light-induced intermittent and
continuous ERK activation in normal rat kidney epithelial cells.
Similar to the TF activity after EGF and NGF stimulation in PC12
cells, intermittent pERK activity caused up-regulation of Fos,
Egfr, Ier2, and Fgf21, which were putatively controlled through
serum response factor (SRF) and CREB binding sites, while
sustained pERK activity caused gene regulation controlled by
AP1 and BACH1. One can speculate that it is more the temporal
dynamics of pERK and less the upstream ligands, such as EGF
or NGF, that eventually encode the transcriptional program
deciding on the cell fate.
To elucidate the various pathways and downstream target
genes under NGF stimulation we constructed a Boolean model
based on our transcriptome and additional literature data.
A prior knowledge network revealed a highly interconnected
pathway map transmitting NGF-induced signals. Training the
network via inhibition of MEK, JNK or PI3K reduced the number
of edges and nodes by about 80% and revealed the MAPK/JNK
pathway as second signaling hub next to MAPK/ERK. Moreover,
blocking the JNK pathway had a more drastic effect on cell
differentiation than blocking MAPK/ERK via inhibition of
MEK through UO126. Indeed, studies on the effect of MEK
inhibition for PC12 cell differentiation are inconclusive. Early
studies report how MEK inhibition completely averted PC12
cell differentiation (Pang et al., 1995; Klesse et al., 1999),
while recent experiments suggest a decrease, rather than full
inhibition of differentiation (Levkovitz et al., 2001; Chung et al.,
2014). Our results were in line with the latter. Despite a
significant reduction in pERK (Figure 5), our cell morphology
measurements detected merely a decrease in the formation of
neurites, rather than full inhibition of differentiation. The reason
for this discrepancy could lie in the time scale of observation.
MEK inhibition delayed differentiation and it took 6 days to
eventually overcome this delay (Figure 4B). This confirmed the
modeling results, which established JNK as key regulator that
is closely interlinked with MAPK/ERK signaling. In concert
with pERK, also pJNK becomes constitutively active upon NGF
stimulation (Figure 2C). Moreover, blocking pERK through
MEK even increased pJNK (and pAKT) levels, while pERK
decreased after JNK inhibition, verifying a crosstalk between JNK
and ERK pathways. Previous reports suggested such a crosstalk
due to dual-phosphatase interaction (Fey et al., 2012), while
other studies proposed that JNK phosphorylates RAF (Adler
et al., 2005; Chen et al., 2012) and thereby contributing to
MAPK/ERK activity. However, the mechanistic details governing
Frontiers in Genetics | www.frontiersin.org
11
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
the crosstalk remain unclear so far. In conclusion, while previous
studies assigned parallel, non-redundant roles to MAPK/ERK
and MAPK/JNK (Waetzig and Herdegen, 2003), our results show
that JNK signaling might be even the main driver for PC12 cell
differentiation.
Next to the negative feedback loops through Klf4, Zfp36, and
Btg2, arresting cell cycle and attenuating mRNA abundance, we
included also two positive feedback loops via uPAR and integrin
signaling as well as through Neuropeptide Y and PKC/PLC
signaling. Positive feedback loops are a common regulatory
pattern in molecular biology to induce bistability switch-like
behavior, particularly in cell fate decisions and differentiation
(Xiong and Ferrell, 2003; Mitrophanov and Groisman, 2008;
Kueh et al., 2013). In fact, multiple feedbacks deciding between
PC12 cell differentiation and proliferation, have been studied on
the level of MAPK signaling (Santos et al., 2007; von Kriegsheim
et al., 2009). Recently, Ryu et al. (2015) used a FRET construct to
quantify pERK dynamics on a single cell level after growth factor
stimulation. While the cell population average still resembled
the hitherto described transient and sustained pERK activity
after respective EGF and NGF stimulation, the authors found
a highly heterogenous response on the single cell level. Pulsed
stimulation, however, not only synchronized MAPK activity
between cells, but also triggered PC12 differentiation upon EGF
stimulation, if the integrated pERK signal was large enough.
The authors concluded that thus not only MAPK signaling, but
also further pathways are responsible for the cell fate decision.
Sparta et al. (2015) used a similar experimental approach to
single cell response of human MCF10A-5e cells to show that
EGFR activity induced a frequency modulation response, while
TrkA activity caused amplitude modulation of pERK levels.
The authors explained these finding by additional receptor-
dependent signaling networks beyond the core Ras-Raf-MEK-
ERK pathway. Extending on this idea, our data and model
suggest autocrine signaling as further feedbacks that sustain the
expression of differentiation inducing TFs. Indeed, uPAR and
also Npy activity were strongly correlated with differentiation
(Figure 3A) and neither Npy nor uPAR signaling were activated
upon EGF stimulation (data not shown). In line with this finding
previous studies reported that uPAR expression is necessary for
NGF-induced PC12 cell differentiation (Farias-Eisner et al., 2000;
Mullenbrock et al., 2011). SERPINE1 regulating the plasminogen
activator-plasmin proteolysis was shown to promote neurite
outgrowth and phosphorylation of the TrkA receptor and ERK
(Soeda et al., 2006, 2008). In our model we included the necessity
of uPAR signaling though the activation of late genes, such as
Klf5, yet the causal relationship between uPAR signaling and
late gene expression remains unclear. However, uPAR signaling
could constitute the additional positive feedbacks beyond MAPK
signaling that were predicted by Ryu et al. (2015), which would
be interesting to test on the single cell level. Reporters for uPAR
and/or JNK activity should likewise show a heterogenous activity
and correlate with the per-cell differentiation status, which could
potentially be modeled within a stochastic differential equation
framework.
In conclusion, our approach has identified the short and
long-term transcriptional activity in PC12 cells after NGF and
EGF stimulation. Modeling the pathway orchestration using a
Boolean model we identified feedback regulations beyond MAPK
signaling that attenuate and sustain the cellular decision toward
differentiation. Extending on previous studies we established JNK
as a key player in PC12 cell differentiation that might have equal,
if not even more importance than ERK during this process.
Over time AP1 was accompanied by a variety of transcription
factors serving signal attenuation, signal maintenance and
morphological change of the cell, which demonstrates that the
decision toward differentiation is a time sequential process over
at least 12 h.
AUTHOR CONTRIBUTIONS
MF, SKn, MK, SK, and BO performed the experiments. MF,
SKn, AS, and BO performed the data analysis. HB And MB
conceived the project, performed the data analysis and wrote the
manuscript with BO. All authors approved the final manuscript.
ACKNOWLEDGMENTS
This
work
was
was
supported
by
the
Deutsche
Forschungsgemeinschaft grant InKoMBio: SPP 1395. The
authors greatly acknowledge the Genomics and Proteomics Core
Facility, German Cancer Research Center/DKFZ, Heidelberg,
Germany for their microarray service.
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found
online
at:
http://journal.frontiersin.org/article/10.3389/fgene.
2016.00044
REFERENCES
Adamski,
D.,
Mayol,
J.-F.,
Platet,
N.,
Berger,
F.,
Hérodin,
F.,
and
Wion, D. (2007). Effects of Hoechst 33342 on C2c12 and PC12 cell
differentiation.
FEBS
Lett.
581,
3076–3080.
doi:
10.1016/j.febslet.2007.
05.073
Adler, V., Qu, Y., Smith, S. J., Izotova, L., Pestka, S., Kung, H. F., et al.
(2005). Functional interactions of Raf and MEK with Jun-N-terminal
kinase
(JNK)
result
in
a
positive
feedback
loop
on
the
oncogenic
Ras
signaling
pathway.
Biochemistry
44,
10784–10795.
doi:
10.1021/
bi050619j
Amit, I., Citri, A., Shay, T., Lu, Y., Katz, M., Zhang, F., et al. (2007). A module
of negative feedback regulators defines growth factor signaling. Nat. Genet. 39,
503–512. doi: 10.1038/ng1987
Angelastro, J. M., Klimaschewski, L., Tang, S., Vitolo, O. V., Weissman, T. A.,
Donlin, L. T., et al. (2000). Identification of diverse nerve growth factor-
regulated genes by serial analysis of gene expression (SAGE) profiling. Proc.
Natl. Acad. Sci. U.S.A. 97, 10424–10429. doi: 10.1073/pnas.97.19.10424
Aoki, K., Kumagai, Y., Sakurai, A., Komatsu, N., Fujita, Y., Shionyu, C., et al.
(2013). Stochastic ERK activation induced by noise and cell-to-cell propagation
regulates cell density-dependent proliferation. Mol. Cell 52, 529–540. doi:
10.1016/j.molcel.2013.09.015
Frontiers in Genetics | www.frontiersin.org
12
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
Avraham, R., and Yarden, Y. (2011). Feedback regulation of EGFR signalling:
decision making by early and delayed loops. Nat. Rev. Mol. Cell Biol. 12,
104–117. doi: 10.1038/nrm3048
Azzalini, A. (2015). The R package sn: The skew-normal and skew-t distributions
(version 1.2-4). Universitate di Padova.
Bar-Joseph, Z., Gitter, A., and Simon, I. (2012). Studying and modelling dynamic
biological processes using time-series gene expression data. Nat. Rev. Genet. 13,
552–564. doi: 10.1038/nrg3244
Bastide, P., Darido, C., Pannequin, J., Kist, R., Robine, S., Marty-Double, C.,
et al. (2007). Sox9 regulates cell proliferation and is required for Paneth cell
differentiation in the intestinal epithelium. J. Cell Biol. 178, 635–648. doi:
10.1083/jcb.200704152
Ben-Dor, A., Shamir, R., and Yakhini, Z. (1999). Clustering gene expression
patterns. J. Comput. Biol. 6, 281–297. doi: 10.1089/106652799318274
Burstein, D. E., Blumberg, P. M., and Greene, L. A. (1982). Nerve growth factor-
induced neuronal differentiation of PC12 pheochromocytoma cells: lack of
inhibition by a tumor promoter. Brain Res. 247, 115–119. doi: 10.1016/0006-
8993(82)91033-2
Cao, X. M., Koski, R. A., Gashler, A., McKiernan, M., Morris, C. F., Gaffney, R.,
et al. (1990). Identification and characterization of the Egr-1 gene product, a
DNA-binding zinc finger protein induced by differentiation and growth signals.
Mol. Cell. Biol. 10, 1931–1939. doi: 10.1128/MCB.10.5.1931
Chao, M. V. (1992). Neurotrophin receptors: a window into neuronal
differentiation. Neuron 9, 583–593. doi: 10.1016/0896-6273(92)90023-7
Chen, J.-Y., Lin, J.-R., Cimprich, K. A., and Meyer, T. (2012). A two-dimensional
ERK-AKT signaling code for an NGF-triggered cell-fate decision. Mol. Cell 45,
196–209. doi: 10.1016/j.molcel.2011.11.023
Chung, J., Kubota, H., Ozaki, Y.-I., Uda, S., and Kuroda, S. (2010). Timing-
dependent actions of NGF required for cell differentiation. PLoS ONE 5:e9011.
doi: 10.1371/journal.pone.0009011
Chung, J., Miura, N., Ito, A., Sawada, M., Nishikawa, S., Kuroda, K., et al.
(2014). Single-cell heterogeneity in suppression of PC12 differentiation by
direct microinjection of a differentiation inhibitor, U0126. Cell Biol. Int. 38,
1215–1220. doi: 10.1002/cbin.10296
Cowley, S., Paterson, H., Kemp, P., and Marshall, C. J. (1994). Activation of
MAP kinase kinase is necessary and sufficient for PC12 differentiation and
for transformation of NIH 3T3 cells. Cell 77, 841–852. doi: 10.1016/0092-
8674(94)90133-3
Dijkmans, T. F., van Hooijdonk, L. W. A., Schouten, T. G., Kamphorst, J. T.,
Fitzsimons, C. P., and Vreugdenhil, E. (2009). Identification of new Nerve
Growth Factor-responsive immediate-early genes. Brain Res. 1249, 19–33. doi:
10.1016/j.brainres.2008.10.050
Dijkmans, T. F., van Hooijdonk, L. W. A., Schouten, T. G., Kamphorst,
J. T., Vellinga, A. C. A., Meerman, J. H. N., et al. (2008). Temporal
and functional dynamics of the transcriptome during nerve growth factor-
induced differentiation. J. Neurochem. 105, 2388–2403. doi: 10.1111/j.1471-
4159.2008.05338.x
Dikic, I., Schlessinger, J., and Lax, I. (1994). PC12 cells overexpressing the insulin
receptor undergo insulin-dependent neuronal differentiation. Curr. Biol. 4,
702–708. doi: 10.1016/S0960-9822(00)00155-X
Eriksson, M., Taskinen, M., and Leppä, S. (2007). Mitogen activated protein kinase-
dependent activation of c-Jun and c-Fos is required for neuronal differentiation
but not for growth and stress response in PC12 cells. J. Cell. Physiol. 210,
538–548. doi: 10.1002/jcp.20907
Farias-Eisner, R., Vician, L., Reddy, S., Basconcillo, R., Rabbani, S. A., Wu, Y. Y.,
et al. (2001). Expression of the urokinase plasminogen activator receptor is
transiently required during “priming” of PC12 cells in nerve growth factor-
directed cellular differentiation. J. Neurosci. Res. 63, 341–346.
Farias-Eisner, R., Vician, L., Silver, A., Reddy, S., Rabbani, S. A., and Herschman,
H. R. (2000). The urokinase plasminogen activator receptor (UPAR) is
preferentially induced by nerve growth factor in PC12 pheochromocytoma cells
and is required for NGF-driven differentiation. J. Neurosci. 20, 230–239.
Fey, D., Croucher, D. R., Kolch, W., and Kholodenko, B. N. (2012). Crosstalk and
signaling switches in mitogen-activated protein kinase cascades. Front. Physiol.
3:355. doi: 10.3389/fphys.2012.00355
Fiore, M., Chaldakov, G. N., and Aloe, L. (2009). Nerve growth factor as a signaling
molecule for nerve cells and also for the neuroendocrine-immune systems. Rev.
Neurosci. 20, 133–145. doi: 10.1515/REVNEURO.2009.20.2.133
Fontanet, P., Irala, D., Alsina, F. C., Paratcha, G., and Ledda, F. (2013).
Pea3 transcription factor family members Etv4 and Etv5 mediate retrograde
signaling and axonal growth of DRG sensory neurons in response to NGF. J.
Neurosci. 33, 15940–15951. doi: 10.1523/JNEUROSCI.0928-13.2013
Gil, G. A., Bussolino, D. F., Portal, M. M., Alfonso Pecchio, A., Renner, M. L.,
Borioli, G. A., et al. (2004). c-Fos activated phospholipid synthesis is required
for neurite elongation in differentiating PC12 cells. Mol. Biol. Cell 15, 1881–
1894. doi: 10.1091/mbc.E03-09-0705
Giraldo, A., Barrett, O. P. T., Tindall, M. J., Fuller, S. J., Amirak, E., Bhattacharya,
B. S., et al. (2012). Feedback regulation by Atf3 in the endothelin-1-responsive
transcriptome of cardiomyocytes: Egr1 is a principal Atf3 target. Biochem. J.
444, 343–355. doi: 10.1042/BJ20120125
Gotoh, Y., Nishida, E., Yamashita, T., Hoshi, M., Kawakami, M., and Sakai,
H. (1990). Microtubule-associated-protein (MAP) kinase activated by nerve
growth factor and epidermal growth factor in PC12 cells. Identity with the
mitogen-activated MAP kinase of fibroblastic cells. Eur. J. Biochem. 193, 661–
669. doi: 10.1111/j.1432-1033.1990.tb19384.x
Greene, L. A., and Tischler, A. S. (1976). Establishment of a noradrenergic clonal
line of rat adrenal pheochromocytoma cells which respond to nerve growth
factor. Proc. Natl. Acad. Sci. U.S.A 73, 2424–2428. doi: 10.1073/pnas.73.7.2424
Györy, I., Boller, S., Nechanitzky, R., Mandel, E., Pott, S., Liu, E., et al.
(2012). Transcription factor Ebf1 regulates differentiation stage-specific
signaling, proliferation, and survival of B cells. Genes Dev. 26, 668–682. doi:
10.1101/gad.187328.112
Hallstrom, T. C., Mori, S., and Nevins, J. R. (2008). An E2f1-dependent gene
expression program that determines the balance between proliferation and cell
death. Cancer Cell 13, 11–22. doi: 10.1016/j.ccr.2007.11.031
Hoffmann, E., Thiefes, A., Buhrow, D., Dittrich-Breiholz, O., Schneider, H., Resch,
K., et al. (2005). MEK1-dependent delayed expression of Fos-related antigen-
1 counteracts c-Fos and p65 NF-kappaB-mediated interleukin-8 transcription
in response to cytokines or growth factors. J. Biol. Chem. 280, 9706–9718. doi:
10.1074/jbc.M407071200
Huang, L., Feng, G., Du, P., Xia, T., Wang, X., Jing, W., et al. (2014). GeneAnswers:
Integrated Interpretation of Genes. R package version 2.10.0.
Ito, E., Sweterlitsch, L. A., Tran, P. B., Rauscher, F. J. III, and Narayanan, R.
(1990). Inhibition of PC-12 cell differentiation by the immediate early gene
fra-1. Oncogene 5, 1755–1760.
Jing, S., Tapley, P., and Barbacid, M. (1992). Nerve growth factor mediates signal
transduction through trk homodimer receptors. Neuron 9, 1067–1079. doi:
10.1016/0896-6273(92)90066-M
Johnson, W. E., Li, C., and Rabinovic, A. (2007). Adjusting batch effects in
microarray expression data using empirical Bayes methods. Biostat 8, 118–127.
doi: 10.1093/biostatistics/kxj037
Kandemir, B., Caglayan, B., Hausott, B., Erdogan, B., Dag, U., Demir, O., et al.
(2014). Pea3 transcription factor promotes neurite outgrowth. Front. Mol.
Neurosci. 7:59. doi: 10.3389/fnmol.2014.00059
Kannan, M. B., Solovieva, V., and Blank, V. (2012). The small MAF transcription
factors MAFF, MAFG and MAFK: current knowledge and perspectives.
Biochim.
Biophys.
Acta
1823,
1841–1846.
doi:
10.1016/j.bbamcr.2012.
06.012
Kaplan,
D.
R.,
Martin-Zanca,
D.,
and
Parada,
L.
F.
(1991).
Tyrosine
phosphorylation and tyrosine kinase activity of the trk proto-oncogene
product induced by NGF. Nature 350, 158–160. doi: 10.1038/350158a0
Klesse, L. J., Meyers, K. A., Marshall, C. J., and Parada, L. F. (1999). Nerve growth
factor induces survival and differentiation through two distinct signaling
cascades in PC12 cells. Oncogene 18, 2055–2068. doi: 10.1038/sj.onc.1202524
Kueh, H. Y., Champhekar, A., Champhekhar, A., Nutt, S. L., Elowitz, M. B.,
and Rothenberg, E. V. (2013). Positive feedback between PU.1 and the
cell cycle controls myeloid differentiation. Science 341, 670–673. doi:
10.1126/science.1240831
Lee, K.-H., Ryu, C. J., Hong, H. J., Kim, J., and Lee, E. H. (2005). CDNA microarray
analysis of nerve growth factor-regulated gene expression profile in rat PC12
cells. Neurochem. Res. 30, 533–540. doi: 10.1007/s11064-005-2688-y
Leppä, S., Saffrich, R., Ansorge, W., and Bohmann, D. (1998). Differential
regulation of c-Jun by ERK and JNK during PC12 cell differentiation. EMBO
J. 17, 4404–4413. doi: 10.1093/emboj/17.15.4404
Levi-Montalcini, R. (1987). The nerve growth factor 35 years later. Science 237,
1154–1162. doi: 10.1126/science.3306916
Frontiers in Genetics | www.frontiersin.org
13
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
Levkovitz, Y., and Baraban, J. M. (2002). A dominant negative Egr inhibitor
blocks nerve growth factor-induced neurite outgrowth by suppressing c-Jun
activation: role of an Egr/c-Jun complex. J. Neurosci. 22, 3845–3854.
Levkovitz, Y., O’Donovan, K. J., and Baraban, J. M. (2001). Blockade of NGF-
induced neurite outgrowth by a dominant-negative inhibitor of the egr family
of transcription regulatory factors. J. Neurosci. 21, 45–52.
Luo, J., West, J. R., Cook, R. T., and Pantazis, N. J. (1999). Ethanol induces
cell death and cell cycle delay in cultures of pheochromocytoma PC12
cells. Alcohol. Clin. Exp. Res. 23, 644–656. doi: 10.1111/j.1530-0277.1999.tb0
4166.x
Luo, W., Friedman, M. S., Shedden, K., Hankenson, K. D., and Woolf, P. J. (2009).
GAGE: generally applicable gene set enrichment for pathway analysis. BMC
Bioinformatics 10:161. doi: 10.1186/1471-2105-10-161
Marek, L., Levresse, V., Amura, C., Zentrich, E., Van Putten, V., Nemenoff,
R. A., et al. (2004). Multiple signaling conduits regulate global differentiation-
specific gene expression in PC12 cells. J. Cell. Physiol. 201, 459–469. doi:
10.1002/jcp.20087
Marshall, C. J. (1995). Specificity of receptor tyrosine kinase signaling: transient
versus sustained extracellular signal-regulated kinase activation. Cell 80, 179–
185. doi: 10.1016/0092-8674(95)90401-8
Mitrophanov, A. Y., and Groisman, E. A. (2008). Positive feedback in cellular
control systems. Bioessays 30, 542–555. doi: 10.1002/bies.20769
Mullenbrock, S., Shah, J., and Cooper, G. M. (2011). Global expression analysis
identified a preferentially nerve growth factor-induced transcriptional program
regulated by sustained mitogen-activated protein kinase/extracellular signal-
regulated kinase (ERK) and AP-1 protein activation during PC12 cell
differentiation. J. Biol. Chem. 286, 45131–45145. doi: 10.1074/jbc.M111.
274076
Murphy, L. O., and Blenis, J. (2006). MAPK signal specificity: the right place at the
right time. Trends Biochem. Sci. 31, 268–275. doi: 10.1016/j.tibs.2006.03.009
Murphy, L. O., MacKeigan, J. P., and Blenis, J. (2004). A network of immediate
early gene products propagates subtle differences in mitogen-activated protein
kinase signal amplitude and duration. Mol. Cell. Biol. 24, 144–153. doi:
10.1128/MCB.24.1.144-153.2004
Murphy, L. O., Smith, S., Chen, R.-H., Fingar, D. C., and Blenis, J. (2002).
Molecular interpretation of ERK signal duration by immediate early gene
products. Nat. Cell Biol. 4, 556–564. doi: 10.1038/ncb822
Müssel, C., Hopfensitz, M., and Kestler, H. A. (2010). BoolNet–an R package for
generation, reconstruction and analysis of Boolean networks. Bioinformatics 26,
1378–1380. doi: 10.1093/bioinformatics/btq124
Pang, L., Sawada, T., Decker, S. J., and Saltiel, A. R. (1995). Inhibition of
MAP kinase kinase blocks the differentiation of PC-12 cells induced by
nerve growth factor. J. Biol. Chem. 270, 13585–13588. doi: 10.1074/jbc.270.
23.13585
Pons, J., Kitlinska, J., Jacques, D., Perreault, C., Nader, M., Everhart, L., et al. (2008).
Interactions of multiple signaling pathways in neuropeptide Y-mediated
bimodal vascular smooth muscle cell growth. Can. J. Physiol. Pharmacol. 86,
438–448. doi: 10.1139/Y08-054
Qui, M. S., and Green, S. H. (1992). PC12 cell neuronal differentiation is associated
with prolonged p21ras activity and consequent prolonged ERK activity. Neuron
9, 705–717. doi: 10.1016/0896-6273(92)90033-A
Ritchie, M. E., Dunning, M. J., Smith, M. L., Shi, W., and Lynch, A. G.
(2011). BeadArray expression analysis using bioconductor. PLoS Comput. Biol.
7:e1002276. doi: 10.1371/journal.pcbi.1002276
Ryu, H., Chung, M., Dobrzynski, M., Fey, D., Blum, Y., Lee, S. S., et al. (2015).
Frequency modulation of ERK activation dynamics rewires cell fate. Mol. Syst.
Biol. 11, 838–838. doi: 10.15252/msb.20156458
Saez-Rodriguez, J., Alexopoulos, L. G., Epperlein, J., Samaga, R., Lauffenburger,
D. A., Klamt, S., et al. (2009). Discrete logic modelling as a means to link protein
signalling networks with functional analysis of mammalian signal transduction.
Mol. Syst. Biol. 5:331. doi: 10.1038/msb.2009.87
Saito, T. H., Uda, S., Tsuchiya, T., Ozaki, Y.-I., and Kuroda, S. (2013). Temporal
decoding of MAP kinase and CREB phosphorylation by selective immediate
early gene expression. PLoS ONE 8:e57037. doi: 10.1371/journal.pone.00
57037
Santos, S. D., Verveer, P. J., and Bastiaens, P. I. (2007). Growth factor-induced
MAPK network topology shapes Erk response determining PC-12 cell fate. Nat.
Cell Biol. 9, 324–330. doi: 10.1038/ncb1543
Sasagawa, S., Ozaki, Y., Fujita, K., and Kuroda, S. (2005). Prediction and validation
of the distinct dynamics of transient and sustained ERK activation. Nat. Cell
Biol. 7, 365–373. doi: 10.1038/ncb1233
Selbie, L. A., and Hill, S. J. (1998). G protein-coupled-receptor cross-talk: the fine-
tuning of multiple receptor-signalling pathways. Trends Pharmacol. Sci. 19,
87–93. doi: 10.1016/S0165-6147(97)01166-8
Shim, K. S., Rosner, M., Freilinger, A., Lubec, G., and Hengstschläger, M. (2006).
Bach2 is involved in neuronal differentiation of N1E-115 neuroblastoma cells.
Exp. Cell Res. 312, 2264–2278. doi: 10.1016/j.yexcr.2006.03.018
Singh, A., Nascimento, J. M., Kowar, S., Busch, H., and Boerries, M.
(2012). Boolean approach to signalling pathway modelling in HGF-
induced
keratinocyte
migration.
Bioinformatics
28,
i495–i501.
doi:
10.1093/bioinformatics/bts410
Soeda, S., Koyanagi, S., Kuramoto, Y., Kimura, M., Oda, M., Kozako, T.,
et al. (2008). Anti-apoptotic roles of plasminogen activator inhibitor-1 as a
neurotrophic factor in the central nervous system. Thromb. Haemost. 100,
1014–1020. doi: 10.1160/th08-04-0259
Soeda, S., Shinomiya, K., Ochiai, T., Koyanagi, S., Toda, A., Eyanagi, R., et al.
(2006). Plasminogen activator inhibitor-1 aids nerve growth factor-induced
differentiation and survival of pheochromocytoma cells by activating both the
extracellular signal-regulated kinase and c-Jun pathways. Neuroscience 141,
101–108. doi: 10.1016/j.neuroscience.2006.03.026
Sparta, B., Pargett, M., Minguet, M., Distor, K., Bell, G., and Albeck, J. G. (2015).
Receptor level mechanisms are required for epidermal growth factor (EGF)-
stimulated extracellular signal-regulated kinase (ERK) activity pulses. J. Biol.
Chem. 290, 24784–24792. doi: 10.1074/jbc.M115.662247
Strickert, M., Teichmann, S., Sreenivasulu, N., and Seiffert, U. (2005). “High-
throughput multi-dimensional scaling (HiT-MDS) for cDNA-array expression
data,” in Artificial Neural Networks: Biological Inspirations ICANN 2005,
number 3696 in Lecture Notes in Computer Science, eds W. Duch, J. Kacprzyk,
E. Oja, and S. A. Zadrrony (Berlin; Heidelberg: Springer), 625–633.
Subramanian, A., Tamayo, P., Mootha, V. K., Mukherjee, S., Ebert, B. L., Gillette,
M. A., et al. (2005). Gene set enrichment analysis: a knowledge-based approach
for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. U.S.A.
102, 15545–15550. doi: 10.1073/pnas.0506580102
Tanabe, K., Bonilla, I., Winkles, J. A., and Strittmatter, S. M. (2003). Fibroblast
growth factor-inducible-14 is induced in axotomized neurons and promotes
neurite outgrowth. J. Neurosci. 23, 9675–9686.
Tiedje, C., Ronkina, N., Tehrani, M., Dhamija, S., Laass, K., Holtmann, H.,
et al. (2012). The p38/MK2-driven exchange between tristetraprolin and HuR
regulates AURich elementdependent translation. PLoS Genet. 8:e1002977. doi:
10.1371/journal.pgen.1002977
Tirone, F. (2001). The gene PC3(TIS21/BTG2), prototype member of the
PC3/BTG/TOB family: regulator in control of cell growth, differentiation, and
DNA repair? J. Cell. Physiol. 187, 155–165. doi: 10.1002/jcp.1062
Vaudry, D., Stork, P. J., Lazarovici, P., and Eiden, L. E. (2002). Signaling pathways
for PC12 cell differentiation: making the right connections. Science 296, 1648–
1649. doi: 10.1126/science.1071552
Vician, L., Basconcillo, R., and Herschman, H. R. (1997). Identification of genes
preferentially induced by nerve growth factor versus epidermal growth factor
in PC12 pheochromocytoma cells by means of representational difference
analysis. J. Neurosci. Res. 50, 32–43.
von Kriegsheim, A., Baiocchi, D., Birtwistle, M., Sumpton, D., Bienvenut, W.,
Morrice, N., et al. (2009). Cell fate decisions are specified by the dynamic ERK
interactome. Nat. Cell Biol. 11, 1458–1464. doi: 10.1038/ncb1994
Waetzig, V., and Herdegen, T. (2003). The concerted signaling of ERK1/2 and
JNKs is essential for PC12 cell neuritogenesis and converges at the level of
target proteins. Mol. Cell. Neurosci. 24, 238–249. doi: 10.1016/S1044-7431(03)
00126-X
Weber, S., Fernandez-Cachon, M. L., Nascimento, J. M., Knauer, S., Offermann,
B., Murphy, R. F., et al. (2013). Label-free detection of neuronal differentiation
in cell populations using high-throughput live-cell imaging of PC12 cells. PLoS
ONE 8:e56690. doi: 10.1371/journal.pone.0056690
Wu, Y. Y., and Bradshaw, R. A. (1996). Synergistic induction of neurite outgrowth
by nerve growth factor or epidermal growth factor and interleukin-6 in PC12
cells. J. Biol. Chem. 271, 13033–13039. doi: 10.1074/jbc.271.22.13033
Xing, J., Kornhauser, J. M., Xia, Z., Thiele, E. A., and Greenberg, M. E.
(1998). Nerve growth factor activates extracellular signal-regulated kinase and
Frontiers in Genetics | www.frontiersin.org
14
April 2016 | Volume 7 | Article 44
Offermann et al.
Boolean Model of PC12 Cell Differentiation
p38 mitogen-activated protein kinase pathways to stimulate CREB serine
133 phosphorylation. Mol. Cell. Biol. 18, 1946–1955. doi: 10.1128/MCB.18.
4.1946
Xiong, W., and Ferrell, J. E. (2003). A positive-feedback-based bistable ‘memory
module’ that governs a cell fate decision. Nature 426, 460–465. doi:
10.1038/nature02089
Yoon, H. S., Chen, X., and Yang, V. W. (2003). Krüppel-like factor 4 mediates p53-
dependent G1/S cell cycle arrest in response to DNA damage. J. Biol. Chem. 278,
2101–2105. doi: 10.1074/jbc.M211027200
Yosef,
N.,
and
Regev,
A.
(2011).
Impulse
control:
temporal
dynamics
in
gene
transcription.
Cell
144,
886–896.
doi:
10.1016/j.cell.2011.
02.015
Zhang, J.-P., Zhang, H., Wang, H.-B., Li, Y.-X., Liu, G.-H., Xing, S., et al. (2014).
Down-regulation of Sp1 suppresses cell proliferation, clonogenicity and the
expressions of stem cell markers in nasopharyngeal carcinoma. J. Trans. Med.
12, 222. doi: 10.1186/s12967-014-0222-1
Conflict of Interest Statement: The authors declare that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2016 Offermann, Knauer, Singh, Fernández-Cachón, Klose, Kowar,
Busch and Boerries. This is an open-access article distributed under the terms
of the Creative Commons Attribution License (CC BY). The use, distribution or
reproduction in other forums is permitted, provided the original author(s) or licensor
are credited and that the original publication in this journal is cited, in accordance
with accepted academic practice. No use, distribution or reproduction is permitted
which does not comply with these terms.
Frontiers in Genetics | www.frontiersin.org
15
April 2016 | Volume 7 | Article 44
|
27148350
|
ERK = ( MEK )
Dusp6 = ( ETS1 )
ECM = ( Mmp3/10 )
BTG2 = ( AKT ) OR ( JNK )
Fosl1 = ( JNK ) OR ( AKT ) OR ( ERK )
MKK7 = ( MEKK1 )
Mmp3/10 = ( Plasmin )
Plasmin = ( uPA/PLAT )
PI3K = ( TrkA )
Npy = ( AP1 )
RAS = ( SOS )
MSK1/2 = ( P38 ) OR ( ERK )
KLF2 = ( AKT ) OR ( JNK ) OR ( ERK )
AKT = ( PI3K )
uPA/PLAT = ( uPAR )
FOS = ( AKT ) OR ( JNK ) OR ( ERK )
JNK = ( MKK7 ) OR ( MEKK4 )
GRB2 = ( SHC )
SHC = ( TrkA ) OR ( FAK )
MKK6 = ( MEKK4 )
DAG = ( PLC )
FRS2 = ( TrkA )
JUNB = ( JNK ) OR ( AKT ) OR ( ERK )
ETS1 = ( JNK ) OR ( ERK )
Egr1 = ( JNK ) OR ( AKT ) OR ( ERK )
RAC1 = ( RAS )
ATF2 = ( P38 ) OR ( JNK ) OR ( ERK )
ZFP36 = ( JNK ) OR ( ERK )
PKC = ( Ca2+ ) OR ( DAG )
P53 = ( AKT ) OR ( JNK ) OR ( ERK )
MEKK4 = ( RAC1 )
KLF6 = ( JNK ) OR ( P53 )
RAF = ( PKC ) OR ( RAS )
KLF4 = ( JNK ) OR ( AKT ) OR ( ERK )
Itga1 = ( ECM )
Mapk3k = ( MEKK4 )
G_i_o = ( NPYY1 )
ARC = ( CREB ) OR ( Egr1 )
SOS = ( GRB2 )
CITED2 = ( CREB ) OR ( ERK ) OR ( P53 )
AP1 = ( JUNB ) OR ( Fosl1 ) OR ( FOS ) OR ( JUND )
KLF10 = ( AKT ) OR ( JNK ) OR ( ERK )
MEKK1 = ( RAC1 )
PLC = ( G_i_o ) OR ( TrkA )
MYC = ( AKT ) OR ( JNK ) OR ( ERK )
SRF = ( RSK )
JUND = ( JNK ) OR ( ERK )
NPYY1 = ( Npy )
P38 = ( Mapk3k ) OR ( MKK6 )
FAK = ( Itga1 ) OR ( RAP1 )
KLF5 = ( AKT ) OR ( ERK ) OR ( P53 )
uPAR = ( AP1 )
Stat3 = ( JNK ) OR ( ERK )
Maff = ( ATF2 ) OR ( JNK ) OR ( ERK )
Ca2+ = ( PLC )
TrkA = ( NGF )
CREB = ( AKT ) OR ( MSK1/2 ) OR ( RSK )
RAP1 = ( C3G )
C3G = ( FRS2 )
MEK = ( RAF ) OR ( MEKK1 )
RSK = ( ERK )
|
RESEARCH ARTICLE
Open Access
Analysis of a dynamic model of guard cell
signaling reveals the stability of signal
propagation
Xiao Gan and Réka Albert*
Abstract
Background: Analyzing the long-term behaviors (attractors) of dynamic models of biological systems can provide
valuable insight into biological phenotypes and their stability. In this paper we identify the allowed long-term
behaviors of a multi-level, 70-node dynamic model of the stomatal opening process in plants.
Results: We start by reducing the model’s huge state space. We first reduce unregulated nodes and simple mediator
nodes, then simplify the regulatory functions of selected nodes while keeping the model consistent with experimental
observations. We perform attractor analysis on the resulting 32-node reduced model by two methods: 1. converting it
into a Boolean model, then applying two attractor-finding algorithms; 2. theoretical analysis of the regulatory functions.
We further demonstrate the robustness of signal propagation by showing that a large percentage of single-node
knockouts does not affect the stomatal opening level.
Conclusions: Combining both methods with analysis of perturbation scenarios, we conclude that all nodes except two
in the reduced model have a single attractor; and only two nodes can admit oscillations. The multistability or oscillations
of these four nodes do not affect the stomatal opening level in any situation. This conclusion applies to the original
model as well in all the biologically meaningful cases. In addition, the stomatal opening level is resilient against single-
node knockouts. Thus, we conclude that the complex structure of this signal transduction network provides multiple
information propagation pathways while not allowing extensive multistability or oscillations, resulting in robust signal
propagation. Our innovative combination of methods offers a promising way to analyze multi-level models.
Keywords: Network model, Discrete dynamic model, Biological network, Signal transduction, Plant signaling, Attractor,
Stomatal opening, Network reduction, Boolean conversion, Stable motif
Background
Modeling offers a comprehensive way to understand bio-
logical processes by integrating the components involved
in them and the interactions between components.
Models can recapitulate and explain the emergent out-
come(s) of the process [1, 2]. Representing cellular pro-
cesses that involve many proteins and small molecules
by a signal transduction network can reveal indirect rela-
tionships between components and provide new insight
[3–5]. Such network usually consists of nodes represent-
ing biological entities, and edges representing interac-
tions. Once a network has been constructed, dynamic
modeling, where each node in the network is associated
with a variable representing its abundance or activity,
can further describe the behavior of the network. Dy-
namic models can have continuous variables whose
change is described by differential equations [6], discrete
variables described by discrete (logical) regulatory func-
tions [7, 8], or a combination of continuous and discrete
variables [9]. The major advantage of discrete dynamic
and continuous-discrete hybrid models is that they use
many fewer parameters than continuous models and
thus need less parameter estimation [10–12]. Modeling
allows one to analyze the biological system represented
by the network in silico, when performing the relevant
experiment is infeasible. It also helps identify general
principles of biological systems [13, 14].
* Correspondence: rza1@psu.edu
Department of Physics, The Pennsylvania State University, University Park, PA,
USA
© 2016 The Author(s). Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Gan and Albert BMC Systems Biology (2016) 10:78
DOI 10.1186/s12918-016-0327-7
The biological process of stomatal opening in plants is
a good example of a complex system wherein modeling
leads to significant gain in understanding [15, 16]. Sto-
mata are pores on leaf surfaces that allow the plant to
exchange carbon dioxide (CO2) and oxygen with the at-
mosphere. Stomata are formed by two guard cells that
can change shape: swelling of guard cells leads to stoma-
tal opening; their shrinking leads to stomatal closure.
The shape of each guard cell is directly controlled by
water flow through the membrane, which is in turn con-
trolled by ion flow. Different signals can affect the guard
cell, changing its ion concentration in direct and indirect
ways, resulting in stomatal opening or closure [17–19].
These signals include light of different wavelengths, CO2
concentration in the air, and plant hormones like absci-
sic acid (ABA). The regulation of stomatal opening is es-
sential to plants, as it controls vital activities like the
uptake of CO2 for photosynthesis, and the unavoidable
water loss through evaporation [20]. Through extensive
experimentation over several decades, more than 70
proteins and small molecules have been identified to
participate in this process.
Sun et al. [15] recently constructed a signal transduc-
tion network based on conclusions from more than 85
articles in the literature, describing how more than 70
nodes (proteins, small molecules, ions) interact with
each other in the stomatal opening process. The net-
work, reproduced as Fig. 1 [15], includes four source
nodes that correspond to the signals red light, blue light,
CO2, and ABA. The more than 150 edges are directed
and signed, with arrowheads indicating activation and
terminal black circles indicating inhibition.
Translating this network into a dynamic model, Sun et
al. characterized each node with a discrete variable de-
scribing its activity and with a discrete (logical) regula-
tory function describing its regulation. Twenty-one out
of the 70 nodes in the model are multi-level, the rest are
Boolean (binary). The levels reflect relative and qualita-
tive information: a level of 2 is a higher level than 1, but
should not be interpreted as twice as high. A few
Fig. 1 The signal transduction network responsible for stomatal opening, as reconstructed by Sun et al. [15]. The color of a node marks which
signal regulates this node. Red nodes are regulated solely by red light. Blue nodes are regulated solely by blue light. Yellow nodes are regulated
solely by ABA. Grey nodes are regulated by CO2. Purple nodes are regulated by both blue and red light. Green nodes are regulated by blue (and
potentially, red) light and ABA. White nodes are source nodes not regulated by any of the four signals. To improve visualization, certain pairs of
edges with the same starting or end nodes overlap. Nodes with multiple levels in the dynamic model are represented by red shadows; the
others are Boolean. The full names of the network components denoted by abbreviated node names are given in Table 1. This figure and part of
its caption is reproduced from Sun Z, Jin X, Albert R, Assmann SM (2014) Multi-level Modeling of Light-Induced Stomatal Opening Offers New
Insights into Its Regulation by Drought. PLoS Comput Biol 10(11): e1003930. doi:10.1371/journal.pcbi.1003930
Gan and Albert BMC Systems Biology (2016) 10:78
Page 2 of 14
discrete values are not integers; e.g. stomatal opening is
a weighted sum with non-integer weights. The dynamic
model has ~1031 states. The logical regulatory functions,
describing each node’s future state based on the states of
the node’s regulators, use a combination of Boolean logic
operators (And, Or, Not), algebraic operations, and
input-output tables. For example, the regulatory function
of PRSL1 is:
PRSL1 ¼ phot1complexOr phot2:
Here for simplicity the node states are denoted by the
node names; the asterisk in “PRSL1*” indicates that this
will be the next state of the PRSL1. The “Or” Boolean
operator expresses that either of the blue light receptors,
i.e. the phot1 complex or phot2, can independently acti-
vate PRSL1.
The Sun et al. model starts from an initial condition
representative of closed stomata. Then a combination of
the four input signals is applied. Red light, blue light,
and ABA are represented as binary variables, and exter-
nal CO2 is represented with three states: 0 (CO2 free
air), 1 (ambient CO2) and 2 (high CO2). The system’s re-
sponse is simulated through repetitive re-evaluation of
each node’s state until a stable value of stomatal opening
is observed. The model successfully captures stomatal
opening in response to combinations of the signals. It
also successfully reproduces stomatal opening under
most of the experimentally studied perturbation scenar-
ios (i.e. genetic knockouts or external supply of compo-
nents). In total, the model is consistent with 63 out of
66 experimental observations collected by Sun et al.
[15]. The model predicts the outcome of a large number
of scenarios that have not been explored experimentally
so far. It also revealed a gap of knowledge regarding the
cross-talk of red light and ABA signaling, and filled it
with a newly predicted interaction.
Although the Sun et al. model recapitulates existing
knowledge and offers new predictions, the model’s full
dynamic repertoire could not be characterized due to its
large state space. Instead, Sun et al. focused on tracking the
output node, stomatal opening, and a few selected internal
nodes, in time. In this paper we apply multiple methods to
analyze the model and aim to fully map all its potential
long-term behaviors, or in other words, attractors.
Methods
Attractors of a dynamical system
An attractor is a set of states from which only states in
the same set can be reached. Attractors that consist of a
single state are called stable steady states or fixed points;
attractors that contain multiple states are called complex
attractors or oscillations [10]. In biological networks,
attractors often have significant biological meaning. In a
cell signaling network, attractors correspond to cell
types, cell fates or behaviors [21]. For example, one at-
tractor can represent a healthy differentiated cell, while
another attractor can represent an abnormally motile
cancer cell [22].
Update scheme of a discrete time model
In the Sun et al. model, as in most discrete dynamic
models, time is an implicit variable. As there is very little
information about the kinetics of the nodes in the sto-
matal opening network, the model incorporates an elem-
ent of stochasticity in timing. The timing does not affect
a system’s fixed point attractors, but it can change the
complex attractors and the possibility of reaching a
given attractor from a given initial state [10]. In the Sun
et al. model, a random–order asynchronous update is
used. Specifically, at each time step, a random order of
nodes (excluding the four input nodes and the output
node stomatal opening) is generated, and each node’s
state is reevaluated in this order; stomatal opening is al-
ways updated last. In the next time step a different order
is generated randomly. In this paper, we use a different
type of stochastic update, called general asynchronous
update, wherein a randomly selected node is updated at
each time step. This is required by the network reduc-
tion method we use. Although this theoretically could
cause a difference in complex attractors, we will show
that in this specific model the two update methods yield
the same attractors.
Network reduction
To reduce the Sun et al. model’s state space, we apply a
network reduction method developed by Saadatpour et
al. [23] that is proven to preserve the attractors of a
Boolean model. Two types of nodes can be reduced
(eliminated or merged): source nodes with no incoming
edges, and simple mediator nodes that have one incom-
ing and/or one outgoing edge. In the reduction, the
source node’s state is directly plugged into the regulatory
function of all of its direct successor nodes; then the
source node is eliminated. For a simple mediator node
with one predecessor (regulator) and one successor (tar-
get), its regulator is connected to its target and the me-
diator node is merged into the regulator. If there is one
regulator and several targets of the mediator node, but
no direct edges between the regulator and any of the tar-
gets, the mediator node is merged into the regulator.
Conversely, if there are several regulators and one target
of the mediator node, but no direct edges among any of
the regulators and the target, the mediator node is
merged into its target. Although this method is not
proven in the multi-level case, we conjecture that attrac-
tors are also conserved for a multi-level model, and will
Gan and Albert BMC Systems Biology (2016) 10:78
Page 3 of 14
show from the results that in the Sun et al. model this
reduction method preserved all attractors.
Elimination of redundant edges
During the process of creating a discrete dynamic model
from biological data, when an influence is weaker than
other influences, the modeler may choose to omit this
influence or, alternatively, include it a redundant way.
The latter choice was made by Sun et al. in four cases,
leading to four regulatory functions that contain an in-
put that does not affect the outcome of the regulatory
function. One of these is
ROS ¼ NADPH And AtrbohD=F Or NADPH
And AtrbohD=F And CDPK Or Not Atnoa1
The italicized words “And”, “Or” and “Not” are Bool-
ean logic operators; the non-italicized words represent
node names. In this regulatory function every node is
Boolean (binary). The first clause “NADPH And Atr-
bohD/F” and the second “NADPH And AtrbohD/F
And CDPK” are connected with an “Or” rule, with the
result that the node “CDPK” does not have any influence
on the outcome. Therefore, we can prune the edge from
CDPK to ROS without changing the model’s dynamics.
We similarly prune three additional redundant edges.
Converting a multi-level model to Boolean
There are several possibilities to convert a multi-level
model to Boolean [24]. The standard method used in the
case of logical models of regulatory networks is the Van
Ham mapping [25, 26]. It preserves the dynamics of the
original model if the variables in the original model can
be represented by integers and if the original model only
allows state transitions in which one node changes its
state by one level [26]. The Sun et al. model does not
satisfy these criteria. However there still is a conclusion
that we can use: All types of conversions maintain the
fixed points and the reachability of states (i.e. if there is
a sequence of state transitions from state A to state B
before conversion, there must be a sequence of state
transitions from the corresponding state A’ to state B’
after the conversion) [26]. So the worst distortion of
attractors due to the conversion is the merging of two
complex attractors into one. In this light we choose to
use an economic mapping of each multi-level node into
as many Boolean nodes as necessary for the binary rep-
resentation of the corresponding integer. We will show
that in this specific model, the conversion did not
change the attractors.
Abbreviations
Table 1 summarizes the full names of the network com-
ponents denoted by abbreviated node names in Fig. 1.
The same abbreviations are used in the original Sun et
al. model and the reduced model developed in this
paper.
Results
Network reduction
The Sun et al. model has a huge state space of ~1031
states, making its analysis difficult. To obtain a smaller
state space, we reduce the size of the network by applying
a network reduction technique developed by Saadatpour
et al. [23] that is proven to preserve the attractors of Bool-
ean models (see Methods). All source nodes other than
the four signals (blue light, red light, CO2, and abscisic
acid) and all simple mediator nodes are identified and re-
duced. This process is done iteratively until it cannot be
done any more. A total of 7 source nodes (14-3-3 protein-
phot1, PIP2C, AtNOA1, Nitrate, PP1cn, mitochondria, and
CHL1), and 19 simple mediator nodes (phot1, phot2,
NIA1, H+-ATPase, LPL, ATP, acid. of apoplast, [NO3
−]v,
[Cl−]v, NADPH, [malate2−]v, PA, ABA receptors, OST1,
PRSL1, PIP2PM, AtrbohD/F, Nitrite, and phot1complex) are
eliminated. Several of the simple mediator nodes form lin-
ear paths (e.g. phot1, OST1) thus their iterative reduction
shortens the linear paths in the network. In addition, 16 of
the 19 reduced mediators have a regulatory function of
the form “B* = A”. It is intuitive that reduction of this node
type preserves the attractors.
We do not eliminate the four signal nodes because we
want to simultaneously explore all the combinations of
input signals. We also choose to not reduce the five
nodes (Kin, Kout, Kc, Ca2+-ATPase, mesophyll cell photo-
synthesis) whose merging with their sole regulator would
result in a self-loop (self-regulation), because such self-
loops may be difficult to interpret. Two additional nodes
with significant biological meaning to the network
(sucrose, stomatal opening), are not reduced either.
Another form of network reduction is the elimination
of redundant edges (see Methods). After removal of re-
dundant edges, the node CDPK becomes a sink node,
thus it can also be eliminated. The reduction of the
above-described nodes and redundant edges simplifies
the network from 70 nodes to 42 nodes, with an esti-
mated state space of ~1022 states.
Simplification of regulatory functions
In order to further reduce the state space from ~1022 to
a manageable size, we grouped state values so that nodes
are represented with fewer states. This grouping was
guided by the 66 experimental observations summarized
in Sun et al.; we aimed to maintain the reduced model’s
results consistent with these experimental observations.
For example, in the Sun et al. model [15] the regula-
tory function of Stomatal Opening is a weighted sum of
different ions and sucrose:
Gan and Albert BMC Systems Biology (2016) 10:78
Page 4 of 14
Stomatal opening ¼ ½Cl−vcontribution
þ ½NO3
−vcontribution þ ½Kþv
þ ½malate2−vcontribution þ sucrose−RIC7=6
The
weights
of
the
anion
contributions
to
the
osmotic potential were chosen based on the literature.
Also, the anion contributions must not exceed a pro-
portion of [K+]v due to charge balance. The anion
contributions are [malate2−]v contribution ≤0.425 × [K+]v;
[NO3
−]v contribution ≤0.10 × [K+]v; [Cl−]v contribution ≤
0.05 × [K+]v. The primary contributions come from [K+]v
and sucrose. We grouped the stomatal opening values into
6 groups with different [K+]v and sucrose values (see
Table 2 and Additional file 1).
The first two columns indicate the [K+]v and su-
crose levels. The third column is the possible values
of stomatal opening in the Sun et al. model for the
given [K+]v and sucrose levels. Note that here we only
show [K+]v, sucrose and stomatal opening value com-
binations
observed
in
the
simulations
of
the
66
Table 1 Full names of the network components denoted by abbreviated node names in Fig. 1
Abbreviation
Full name
Abbreviation
Full name
14-3-3
proteinH-
ATPase
14-3-3 protein that binds to the H+-ATPase
14-3-3
proteinphot1
14-3-3 protein that binds to phototropin 1
ABA
abscisic acid
ABI1
2C-type protein phosphatase
acid. of
apoplast
the acidification of the apoplast
AnionCh
anion efflux channels at the plasma
membrane
AtABCB14
ABC transporter gene AtABCB14
Atnoa1
protein nitric oxide-associated 1
AtrbohD/F
NADPH oxidase D/F
AtSTP1
H-monosaccharide symporter gene AtSTP1
Ca2+-ATPase
Ca2+-ATPases and Ca2+/H+ antiporters responsible for Ca2+ efflux from
the cytosol
CaIC
inward Ca2+ permeable channels
CaR
Ca2+ release from intracellular stores
carbon fixation
light-independent reactions of
photosynthesis
CDPK
Ca2+-dependent protein kinases
CHL1
dual-affinity nitrate transporter gene
AtNRT1.1
Ci
intercellular CO2 concentration
FFA
free fatty acids
H+-ATPase
the phosphorylated H+-ATPase at the plasma membrane prior to the
binding of the H+-ATPase 14-3-3 protein
H
+-ATPasecomplex
14-3-3 protein bound H+-ATPase
KEV
K+ efflux from the vacuole to the cytosol
Kin
K+ inward channels at the plasma
membrane
Kout
K+ outward channels at plasma membrane
LPL
lysophospholipids
NADPH
reduced form of nicotinamide adenine dinucleotide phosphate
NIA1
nitrate reductase
NO
nitric oxide
OST1
protein kinase open stomata 1
PA
phosphatidic acid
PEPC
phosphoenolpyruvate carboxylase
phot1
phototropin 1
phot1complex
14-3-3 protein bound phototropin 1
phot2
phototropin 2
Photophos-
phorylation
light-dependent reactions of
photosynthesis
PIP2C
phosphatidylinositol 4,5-bisphosphate located in the cytosol
PIP2PM
phosphatidylinositol 4,5-bisphosphate
located at the plasma membrane
PLA2β
phospholipase A2β
PLC
phospholipase C
PLD
phospholipase D
PMV
electric potential difference across the
plasma membrane
PP1cn
the catalytic subunit of type 1 phosphatase located in the nucleus
PP1cc
the catalytic subunit of type 1 phosphatase
located in the cytosol
protein
kinase
a serine/threonine protein kinase that directly phosphorylates the
plasma membrane H-ATPase
PRSL1
type 1 protein phosphatase regulatory
subunit 2-like protein1
RIC7
ROP-interactive CRIB motif-containing protein 7
ROP2
small GTPase ROP2
ROS
reactive oxygen species
[Ca2+]c
cytosolic Ca2+ concentration
[Cl−]c/v
cytosolic/vacuolar Cl−concentration
[K+]c/v
cytosolic/vacuolar K+ concentration
[malate2−]a/c/v
apoplastic/ cytosolic/vacuolar malate2−concentration
[NO3
−]a/c/v
apoplastic/cytosolic/vacuolar nitrate
concentration
Gan and Albert BMC Systems Biology (2016) 10:78
Page 5 of 14
experimentally studied scenarios reported by Sun et
al. [15]. More stomatal opening values are possible
when considering node perturbations. The 4th col-
umn shows the simplified stomatal opening level after
grouping. The update function for the simplified sto-
matal opening level covers all possible values of [K+]v
and sucrose (see Additional file 1).
Similarly to the original model, the simplified states
represent qualitative, relative categories. For example, a
stomatal opening level of 2 is not twice as high as level
1. We choose the simplified stomatal opening values so
that there is no state “4”, to better reflect an experimen-
tally observed synergistic effect between blue and red
light [18, 19, 27]. Simulation results with the simplified
regulatory function are that under monochromatic red
light stomatal opening =1; under monochromatic blue
light stomatal opening =3; under dual beam the stomatal
opening =5, which is larger than the sum “1 + 3”. This
qualitatively reproduces the experimental observation
that under dual beam illumination stomata open to a
size much larger than the sum of opening under mono-
chromatic blue or red light.
We find by simulation of the reduced model, using
the same initial condition as the Sun et al. model,
that the simplification of the stomatal opening regula-
tory function results in only 3 additional cases of in-
consistency with experimental observations out of a
total of 66 experimentally studied scenarios. Add-
itional file 2 lists all experimental observations and com-
pares them to the relevant simulation results. Ignoring the
contribution of malate2−, NO3
−, and RIC7 to stomatal open-
ing each causes one additional discrepancy; ignoring Cl−
does not cause any additional discrepancy. Ignoring these
nodes trades a decrease in accuracy for a significant in-
crease in simplicity.
The simplification of the stomatal opening regulatory
function eliminates the effect of vacuolar anions and of
RIC7 on stomatal opening. As a result we can further
simplify the Sun et al. model by eliminating 10 nodes in
total, [malate2−]a, [malate2−]c, starch, [Cl−]c, [NO3
−]c,
[NO3
−]a, ROP2, RIC7, ABC, and PEPC. The only edge
from these nodes to other nodes is [malate2−]a →
AnionCh. In section 3 of Additional file 3 we show that
eliminating this edge does not change the system’s long-
term behavior, i.e. attractors. Also, the regulatory func-
tion describing the cytosolic K+ concentration, [K+]c, can
be simplified without loss, as described in section 3 of
Additional file 3. After this simplification we have a net-
work of 32 nodes, 81 edges, indicated on Fig. 2. We will
refer to this model as the “reduced model”. A list of
nodes and their regulatory functions is provided in
Additional file 1.
Identifying strongly connected components (SCCs)
is important for attractor analysis, as complex dy-
namic behavior such as oscillations or multi-stability
requires feedback loops [7]. There are three SCCs in
the network of the reduced model, as marked in
Fig. 2. The NO cycle contains three nodes and three
positive edges. The Ci SCC contains three nodes,
which form two negative feedback loops. The Ion
SCC is the most complex, containing 13 nodes and
26 edges, 7 of which are negative.
Next
we
perform
attractor
analysis
using
two
methods: 1. by converting the reduced model to Boolean
and applying two analysis tools; 2. by analyzing the regula-
tory functions theoretically. The former method finds all
stable steady states and candidate oscillations; the latter
confirms the results of the first method and gives insight
about perturbation scenarios.
Conversion of nodes from multi-level to Boolean states
and attractor analysis
We perform the conversion to Boolean to enable at-
tractor analysis by existing software tools. Zañudo et al.
[28] proposed an algorithm to find the attractors of a
Boolean network based on the concept of “stable motif”,
a strongly-connected group of nodes that can stabilize
regardless of their inputs. The algorithm finds all stable
motifs, which determine the part of the network that
stabilizes in an attractor. After a stable motif is found,
one can plug in its stabilized state into the network, and
obtain a smaller remaining network. After repeating this,
eventually the remaining part is either nothing (indicat-
ing a fixed point/stable steady state) or a candidate oscil-
lating sub-network. Compared with other software tools
[29, 30], the major advantage of this algorithm is that it
finds all the attractors of Boolean networks with hun-
dreds of nodes [28]. Application of this powerful method
requires a Boolean model, so we convert the multi-level
model into Boolean first (see Methods). An example of
conversion is given in Table 3.
Table 2 Grouping of the stomatal opening values by the level
of [K+]v and sucrose
[K+]v
Sucrose
Stomatal opening value
in the Sun et al. model
Simplified stomatal
opening value
0
0
0
0
0
1 or 2
1 or 2
1
1
0
1.58
1
1.8
1
3.84
2
1.5
2
4.36
2
2
0 or 1
3.15 or 4.15
3
4.5
0 or 2
5.18 or 8.92
3
6
0
9.28 or 9.45
5
6
2
11.28 or 11.45
5
9
0 or 2
14.01 or 16.01
6
Gan and Albert BMC Systems Biology (2016) 10:78
Page 6 of 14
More detailed examples of the conversion of the states
and regulatory function of specific nodes are given in
the Additional file 4. We will refer to the reduced model
after conversion to Boolean variables as the “Boolean-
converted reduced model”. The regulatory functions of
the Boolean-converted reduced model are available in
Additional
file
5.
When
simulating
the
Boolean-
converted reduced model, all the Boolean nodes that
Fig. 2 The stomatal opening network after model reduction, with 32 nodes and 81 edges. Nodes with shadows have multiple states; other nodes
are binary. The three strongly-connected components (SCCs) of the network are indicated by rectangles with dashed contours
Gan and Albert BMC Systems Biology (2016) 10:78
Page 7 of 14
represent the same entity (the same multi-level node)
are updated simultaneously. In this way the state transi-
tions of the reduced model will be kept the same in the
Boolean-converted reduced model, and therefore the
Boolean conversion will not cause additional discrepan-
cies from experimental observations.
We apply the stable motif algorithm’s implementation,
downloaded
from
http://github.com/jgtz/StableMotifs/
[28], to the Boolean-converted reduced model. The algo-
rithm uses the Boolean regulatory functions of the con-
verted model (given in Additional files 5 and 6) as input.
We consider every combination of sustained states of
the five signal nodes (blue light, red light, ABA, CO2,
CO2_high). We find two possible stable motifs, corre-
sponding to the self-regulatory node PMV_pos (one of
the two Boolean nodes associated with the multi-level
node PMV, see Additional files 4 and 5), in conditions
where the H+-ATPasecomplex is inactive. These two stable
motifs indicate the bistability of PMV. Under its influ-
ence, another node, Kout, will also be bistable. The algo-
rithm also indicates that for any signal combination,
every
node,
except
[Ca2+]c
and
Ca2+-ATPase,
will
stabilize in a fixed state. [Ca2+]c has three states, and in
the Boolean-converted model it is represented by two
nodes, Cac and Cac_high. Cac_high, which represents
the higher level of [Ca2+]c, stabilizes at zero in all situa-
tions. Cac and Ca2+-ATPase may oscillate in conditions
where blue light is present and ABA is absent (a total of
six cases, two of which allow PMV bistability). Table 4
summarizes key features of the attractors found by the
stable motif algorithm for all 24 input combinations.
Attractors where Ca2+ oscillation is not possible are
fixed points (stable steady states).
We verified the obtained attractors with GINsim [12],
a software suite capable of model construction, simula-
tion, and analysis. GINsim can compute all stable steady
states (called stable states in GINsim), or determine
complex attractors by mapping the state transitions. The
stable steady states found by GINsim are identical to
those found by the stable motif algorithm. To verify and
further explore the complex attractors, we use the simu-
lation function of GINsim, starting from a state in the
complex attractor. The result that the system oscillates
between four states, where only the state of Cac and Ca2
+-ATPase changes, agrees with the findings of the stable
motif algorithm. We summarize the GINsim computa-
tion/simulation results in Additional file 7. Additional
file 8 indicates the Boolean-converted reduced model in
SBML-qual format [29], a general format for biological
model to be analyzed using various tools including
GINsim.
We can also connect the stable motif analysis results
to network reduction. We have previously decided to
not reduce the four nodes that correspond to input sig-
nals. If we do consider a specific input combination
when using network reduction, e.g. blue light and red
light with normal CO2 without ABA, we can reduce
much more of the network: two of the three SCCs,
namely the NO cycle and the Ci SCC, will stabilize and
can be eliminated. Only the Ion SCC and its sole output
stomatal opening remain, indicating that this SCC is not
driven solely by the external signals and has the capacity
for oscillations or multi-stability. This is consistent with
the results found by stable motif analysis, according to
which the NO cycle and the Ci SCC attain a steady state
and the Ion SCC admits a [Ca2+]c - Ca2+-ATPase oscilla-
tion and PMV bistability. This consistency supports the
appropriateness of the network reduction method and of
the Boolean conversion.
Theoretical analysis of the reduced model
To gain additional insight into the attractors of the re-
duced model and their potential changes due to node
perturbations, we analyze the reduced model theoretic-
ally. Specifically, we aim to answer the question: Can
there be other types of oscillation, or can there be add-
itional multi-stability, if a node is knocked out (fixed in
the OFF state) or is constitutive active (fixed in the high-
est state)?
We first test whether the network and regulatory rules
allow multi-stability or oscillations. This analysis is based
on R. Thomas’s conjectures [7]: The presence of a positive
(negative) feedback loop - a cycle with an even (odd) num-
ber of inhibitory edges - in the network is a necessary but
not sufficient condition for the occurrence of multiple
steady states (oscillations). The conjectures have been
proven in the case of discrete dynamic systems [31–34].
Since only feedback loops are candidates for potential
multi-stability or oscillations, we analyze the regulatory
functions of each strongly connected component of the
network. For each feedback loop, we identify a sufficient
condition for the nodes to stabilize in a specific state. The
violation of this condition becomes a further necessary
condition of multi-stability or oscillation. Here we de-
scribe the main steps and results of the analysis; the de-
tailed analysis is in Additional file 3.
Table 3 Example of Boolean conversion
Level of the original
node
State of Boolean
node_2
State of Boolean
node_1
0
0
0
1
0
1
2
1
0
3
1
1
The multi-level node shown in the 1st column is mapped into two Boolean
nodes, shown in the 2nd and 3rd columns, using the binary representation of
the corresponding integer.
Gan and Albert BMC Systems Biology (2016) 10:78
Page 8 of 14
The NO cycle is composed of the nodes PLD, ROS,
NO, and the three positive edges between them. It does
not have any negative edges, so it cannot oscillate. A
fixed ABA value is sufficient to stabilize each node of
the cycle in a specific state, thus the cycle does not
admit multi-stability under any perturbation.
The Ci SCC has three nodes, Ci, mesophyll cell photo-
synthesis (MCPS), carbon fixation, and four edges that
form two negative feedback loops, one between carbon
fixation and Ci, and the other between Ci and MCPS.
Despite the existence of negative feedback, this cycle will
stabilize if given a fixed CO2 value. From this we know
that this cycle cannot oscillate or admit multi-stability
under any perturbation.
The Ion SCC has 13 nodes. To reduce its complexity
we show that the key node [Ca2+]c, which has states 0,1,
and 2, cannot enter state 2 in the long term under any
perturbation. Since most nodes respond to [Ca2+]c only
if [Ca2+]c =2, we can eliminate all edges that depend
only on “[Ca2+]c =2”, and obtain a simplified Ion SCC,
as shown in Fig. 3. The Ca2+ SCC ([Ca2+]c, Ca2+ ATPase,
PLC, CaR) now becomes a sink SCC. The only negative
edge in this sub-network is from Ca2+-ATPase to [Ca2+]c.
These two nodes are known to oscillate. The positive feed-
back loop formed by [Ca2+]c, PLC, and CaR will stabilize if
given fixed inputs. So there cannot be multi-stability. For
the nodes outside of the Ca2+ feedback loops, we show that
the edges from KEV and [K+]v are redundant in the long
term, so there are no feedback loops except the PMV self-
loop. PMV is not capable of having oscillations, but can
have bistability (as also indicated by the stable motif
analysis). The bistability can affect at most one other node,
Kout, under any perturbation. This means that the bistability
has very limited effect on the attractor of the reduced
model.
Now we can summarize our conclusions and return to
the question we sought to answer: there is no oscillation
except in the calcium nodes; there is no multi-stability
except in the nodes PMV and Kout. These statements are
true under any perturbation. Moreover, for the calcium
oscillation, [Ca2+]c cannot enter the state 2, so the sub-
network between [Ca2+]c and Ca2+-ATPase is a negative
feedback loop between two Boolean nodes, with the
regulatory functions Ca2+ ATPase* = [Ca2+]c; [Ca2+]c* =
not Ca2+ ATPase. It results in the simplest type of oscil-
lation, as also found by GINsim simulation. For the
PMV bistability, even if the bistability exists, most nodes,
especially the output node stomatal opening, still have a
unique value. Thus the theoretical analysis, in agreement
with the computational analysis, leads to very strong
conclusions
about
the
reduced
model’s
dynamic
repertoire.
We can also show that the reduction or Boolean con-
version did not change the attractors of the Sun et al.
model. Although the reduction we used is only proven
in the Boolean case, Naldi et al. showed that for multi-
valued models, removal of non-autoregulated nodes, like
in our reduction, preserves crucial dynamical properties
[35], including fixed point attractors and the two-node
simple oscillation we found. So our reduction is valid in
this specific model. To confirm that the Boolean conver-
sion preserved attractors, we note that in the Boolean-
Table 4 Summary of the attractors found using the stable motif algorithm
BL
RL
CO2
CO2_high
ABA
SO (Bool)
SO
Ca2+ Oscillation Possible?
PMV_pos bistability
0
0
Any
Any
Any
000
0
No
Yes
0
1
0
0
1
000
0
No
No
0
1
1
Any
1
000
0
No
Yes
1
Any
1
0
1
000
0
No
No
1
Any
1
1
1
000
0
No
Yes
0
1
1
Any
0
010
1
No
Yes
1
Any
1
1
0
010
1
Yes
Yes
0
1
0
0
0
101
3
No
No
1
0
1
0
0
101
3
Yes
No
1
Any
0
0
1
101
3
No
No
1
0
0
0
0
110
5
Yes
No
1
1
1
0
0
110
5
Yes
No
1
1
0
0
0
111
6
Yes
No
The first five columns indicate the input signal combination. The setting CO2_high = 1 and CO2 = 0 is not included because it is not biologically meaningful. The
“SO (Bool)” column indicates the state of the Boolean node combination representing stomatal opening. The “SO” column is the state of stomatal opening when
converted back to an integer. Note that the stomatal opening level of four is not defined, and no attractors have a stomatal opening level of two. The next
column indicates whether Ca2+ oscillation can possibly happen under the given signal combination. The last column indicates whether bistability of PMV_pos can
be observed under this setting. In those cases, two stable steady states with (PMV_pos = 0, Kout = 0) and (PMV_pos = 1, Kout = 1) can be observed. The rest of the
nodes are unaffected by this two-node bistability
Gan and Albert BMC Systems Biology (2016) 10:78
Page 9 of 14
converted reduced model we found fixed point attractors
and a complex attractor in which only two nodes oscil-
late. Because the only potential change to attractors as a
consequence of the conversion is merging of complex
attractors [26], it is straightforward that the attractors
have been conserved during the conversion, as the two-
node oscillation found is the simplest type of complex
attractor and cannot be a result of attractor merging. In
addition, using general asynchronous update instead of
random order asynchronous update does not cause any
changes to the attractor, because the update schemes do
not affect fixed points or the two-node simple oscillation
we found.
Stability of guard cell signal transduction
Our previous results indicate the stability of the system
in the sense that all the initial conditions lead to the
same attractor except for up to four nodes. We also
examine another facet of the system’s stability: the ro-
bustness of the stomatal opening in response to node
perturbations that render them non-functional. We per-
form a systematic analysis of single-node knockouts of
every non-signal node in the reduced model, under all
combinations of light, CO2 and ABA conditions. For
each signal combination, we set the perturbed node’s ini-
tial state and regulatory function to 0, initialize the rest
of the nodes in the condition representative of closed
stomata, and then simulate the reduced model until it
reaches its attractor. In the absence of ABA under each
light and CO2 condition, 60–90 % perturbation scenarios
produce the same stomatal opening value as the unper-
turbed system (Table 5). These results are similar to
those reported by Sun et al. for the original model [15]
(see Additional file 9). In the presence of ABA 50–90 %
perturbation scenarios produce the same stomatal open-
ing value as the unperturbed system, and 4–16 % knock-
outs
lead
to
a
higher
stomatal
opening
value.
Perturbations in the ABA = 1 case were not studied by
Sun et al., but our simulations of the original model give
the same qualitative results as the reduced model. These
results indicate the closeness of the perturbed attractor
(at least in terms of the stomatal opening value) to the
unperturbed attractor in more than 50 % of single node
perturbations. They also suggest the resilience of the sto-
matal opening process against internal failures and
perturbations.
Extending the conclusions to the original model
We found that in the reduced model there is no oscilla-
tion except in the calcium nodes; there is no multi-
stability except in the nodes PMV and Kout. Because the
reduction
we
used
has
been
shown
to
conserve
Fig. 3 The Ion SCC after reducing all edges that depend on calcium. All regulators of this sub-network have been omitted. On the left, [Ca2+]c
related nodes form a sink sub-network
Gan and Albert BMC Systems Biology (2016) 10:78
Page 10 of 14
attractors [23, 35], we know that our attractor conclu-
sions can be immediately extended to all nodes in the
original model except the reduced nodes and stomatal
opening. Next we extend the attractor analysis to include
the reduced nodes as well.
First we consider the nodes reduced during the first
step of network reduction, i.e. non-signal source nodes
and simple mediator nodes. These nodes are trivially in-
capable of having multi-stability and oscillations them-
selves, so we need only to consider their perturbations.
Perturbation of a simple mediator node can always be
replaced by a corresponding (set of) perturbation(s) in
the mediator node’s direct successor(s), so these pertur-
bations have already been considered. Perturbing a non-
signal source node may theoretically cause a difference,
however the nodes in this category in the Sun et al.
model represent molecules that are abundant in the cell
or cell environment, thus their perturbation is not bio-
logically relevant or practical.
Next we consider the anion nodes reduced due to the
simplified stomatal opening rule. Recall that these nodes
do not affect other nodes except stomatal opening in the
long term. There cannot be multi-stability in anion
nodes unless the assumptions of sufficient initial [NO3
−]a
and starch concentration, and sufficient initial mito-
chondrial TCA cycle activity are violated (details are
provided in Additional file 3, section 5 and 6). Since
there is no support for interventions that would lead to
the violation of these assumptions, it is reasonable to
conclude that no multi-stability can be found in the re-
duced nodes under biologically relevant situations. We
also found that there can be an additional oscillation in
the RIC7 path (involving the nodes ROP2, RIC7 and SO)
when a special set of perturbations is applied. Under that
case, the nodes RIC7 and SO will oscillate. Since the ef-
fect of this behavior is small (within 5 % of the unper-
turbed SO value in the Sun et al. model [15]), it has
little biological significance. There are no more possible
oscillations as there are no more negative feedback
loops. To conclude, the original Sun et al. model has os-
cillations only in cytosolic Ca2+ ([Ca2+]c) and Ca2+
ATPase, and has multi-stability only in PMV and Kout,
under situations that are biologically meaningful.
Discussion
The conclusions we obtained can tell us how to control
this network model. Generally in engineering applica-
tions, control means to drive a system into an arbitrary
state [36, 37]. However in biological systems, it is more
meaningful to drive the system into one of its natural
attractors rather than into an arbitrary state, as the
attractors correspond to stable phenotypes [38]. To con-
trol the attractor of a Boolean system, one needs to con-
trol only its input nodes and a subset of nodes in each
Table 5 Summary of systematic perturbation results
Light, CO2 and ABA condition
Unperturbed
SO level
Simplified SO level
Percentage
of cases
with
unchanged
SO value
0
1
2
3
5
6
Percentage of single knockouts that lead to each SO level
Dual Beam
Mod. CO2
ABA OFF
5
4 %
31 %
65 %
65 %
Low CO2
6
31 %
4 %
65 %
65 %
High CO2
1
4 %
96 %
96 %
Blue Light
Mod. CO2
3
35 %
65 %
65 %
Low CO2
5
31 %
4 %
65 %
65 %
High CO2
1
4 %
96 %
96 %
Red Light
Mod. CO2
1
4 %
96 %
96 %
Low CO2
3
35 %
65 %
65 %
High CO2
1
4 %
96 %
96 %
Dual Beam
Mod. CO2
ABA ON
0
85 %
4 %
8 %
4 %
85 %
Low CO2
3
46 %
50 %
4 %
50 %
Blue Light
Mod. CO2
0
85 %
4 %
8 %
4 %
85 %
Low CO2
3
46 %
50 %
4 %
50 %
Red Light
Low CO2
0
96 %
4 %
96 %
The first set of columns, with the header ‘Light, CO2 and ABA condition’, indicate the input signal combinations. The abbreviation “Mod.” means moderate CO2
concentration. Note that we do not list the four input combinations (high CO2 with ABA and with any type of light, or moderate CO2 with ABA and red light)
wherein all simulated stomatal opening values are zero. The 2nd column is the simulated stomatal opening (SO) level in the unperturbed system. The 3rd column
set shows the percentage of single-node knockouts that yield the corresponding SO level. There is no stomatal opening level 4 in the reduced model. No entry
means zero percentage. The last column is the percentage of settings where the stomatal opening remains at the same level as the unperturbed case. A complete
table of perturbation results is provided in Additional file 9
Gan and Albert BMC Systems Biology (2016) 10:78
Page 11 of 14
stable motif [39]. Our integrated analysis, involving
Boolean conversion, indicates that to control the at-
tractor that the stomatal opening network evolves into,
one only needs to control the input signals and PMV,
even in case of perturbations. In particular, to control
the stomatal opening value, one only needs to control
the input signals, under any perturbation.
The reduced model provides new biological insights.
Normally, when ABA is present, stomata will close.
However in
some knockout
mutants stomata can
open to a certain extent in the presence of ABA, al-
though the opening level is not as much as in the
case without ABA [15]. Such partial reversals of the
effect of ABA are important for understanding the
mechanism of stomatal opening. For example, Sun et
al. reported that OST1 knockout (OST1 is kept 0)
and inhibition of the NADPH oxidase (AtrbohD/F is
kept 0) yielded partially restored SO level in simula-
tions, in agreement with experimental observations
(see Additional file 2 for the comparison of the
equivalent simulations in the reduced model with
experiments). Simplification of the Sun et al. model
allows easier simulation of more perturbation scenar-
ios, e.g. the systematic identification of possible par-
tial reversals. Table 6 indicates all the partial reversals
due to single node knockouts in the reduced model.
Our results reproduce the observation that knockout
of nodes in the ABA pathway (PLD, NO, ROS) can cause
partial reversals of ABA’s effect. We find that AnionCh
knockout can partially restore stomatal opening inhib-
ited by ABA, a result not reported by Sun et al., but
which is supported by experimental evidence [40]. In
addition, Table 6 offers a new biological prediction: low
CO2 concentration can partially restore stomatal open-
ing when ABA is present. This is consistent with the
knowledge that CO2-free air promotes stomatal opening
in the absence of ABA [41]. This CO2 effect suggests a
mechanism of cross-talk between CO2 and ABA. Im-
portantly, apart from the five nodes listed in Table 6, no
other node’s knockout can reverse ABA’s inhibition of
stomatal opening. The perturbation results of Table 5
offer many more new predictions.
Our combination of techniques offers a powerful frame-
work for determining the dynamic repertoire of a multi-
level dynamic model. Multi-level models are more accur-
ate than Boolean models in describing the quantitative
characteristics of dynamic systems, but there are few gen-
eral methods to analyze multi-level models [10, 12]. By
combining different existing methods, we were able to
overcome the limitations of each method. Our successful
combination of existing methods offers a promising way
to analyze multi-level models, and might point towards a
general strategy to analyze the attractors of multi-level
models, biological or non-biological.
A notable future direction for this work is to develop
an alternative way to determine the attractors of multi-
level models by extending the concept of stable motifs.
Compared with conversion to a Boolean model, then ap-
plying Boolean stable motif algorithm, extending the
stable motif algorithm to multi-level models can avoid
potential attractor change issues. Development of such a
technique will allow easy and powerful attractor analysis
for multi-level models.
Conclusions
We obtained a very strong conclusion about the attrac-
tors of the Sun et al. stomatal opening model: under any
combination of sustained signals, all nodes in the model
converge into steady states, with the potential exception
of the cytosolic Ca2+ ([Ca2+]c) and Ca2+ ATPase. Varia-
tions in the initial condition of non-source nodes or in
process timing (node update sequence) can drive at most
two nodes, PMV and Kout, into a different attractor. This
high degree of attractor similarity is somewhat unex-
pected, as the network has a large strongly connected
component and several feedback loops. Thus, despite the
decidedly non-linear structure of the network, most parts
of the system behave in the consistent manner of a linear
pathway. This is a distinct feature of the stomatal opening
model: many dynamic models of biological systems have
multiple, diverse attractors [22, 42]. The models of these
systems will evolve into drastically different attractors
when starting from different initial conditions, sometimes
even when starting from the same initial condition, dem-
onstrating different biological trajectories. In the stomatal
opening model, however, the uniqueness of the steady
state stomatal opening level suggests that the final extent
of the stomatal opening response is robust and resilient
Table 6 Nodes whose knockouts diminish ABA’s inhibition of stomatal opening
Light, CO2 and ABA condition
Unperturbed
SO level
Nodes whose knockout results in a partially restored SO, and the corresponding SO value
CO2
NO
PLD
ROS
AnionCh
Dual Beam
Moderate CO2, ABA is present
0
3
3
5
3
2
Blue Light
0
3
2
3
2
1
Red Light
0
3
1
The first set of columns, with the header ‘Light, CO2 and ABA condition’, indicate the input signal combinations. The 2nd column is the stomatal opening without
perturbations. The 3rd column set indicates the nodes whose knockout would yield a stomatal opening level that is higher than the unperturbed value of 0. CO2
knockout means CO2 being set to zero (CO2 free air). No entry means the setting does not cause partial reversal
Gan and Albert BMC Systems Biology (2016) 10:78
Page 12 of 14
against changes in initial conditions or in timing. Note
that although a change in the initial condition will not
change the steady-state opening level, it may change the
steady state of PMV and Kout, and may change how fast
the system converges to an attractor.
We also showed that the reduced stomatal opening
model does not admit additional, emergent oscillations or
multi-stability under any biologically relevant node per-
turbation (knockout or constitutive activity). We further
demonstrate the robustness of the system by examining
the stomatal opening level under single node knockouts: in
most cases the signals are still likely to propagate and lead
to a similar degree of stomatal opening as in the absence of
perturbation. This robustness is unlike a single linear path-
way, which would be very sensitive to node disruption. We
suggest that the role of the strongly connected components
in the network could be to provide multiple paths for the
signal to propagate, but at the same time not allowing ex-
tensive multistability or oscillations. Our innovative com-
bination of existing methods offers a promising way to
analyze multi-level models.
Additional files
Additional file 1: Regulatory functions of the reduced stomatal opening
model. (DOCX 38 kb)
Additional file 2: Compilation of comparisons between published
experimental observations and the reduced model’s results for
simulations of the identical conditions. (DOCX 163 kb)
Additional file 3: Analysis of stomatal opening model. Detailed
derivation of attractor analysis and other statements made in the main
article. (DOCX 98 kb)
Additional file 4: Examples of converting a multi-level update function
to Boolean. (DOCX 38 kb)
Additional file 5: Regulatory functions of the Boolean-converted reduced
stomatal opening model. (DOCX 25 kb)
Additional file 6: Text file of the Boolean-converted reduced stomatal
opening model to be used in the stable motif algorithm. The stable motif
algorithm and instructions about how to use it can be found from this
link: https://github.com/jgtz/StableMotifs. (TXT 4 kb)
Additional file 7: GINsim attractor analysis of the Boolean-converted
reduced stomatal opening model. (DOCX 42 kb)
Additional file 8: Boolean-converted reduced stomatal opening model
in SBML format. This file format can be used in various tools, including
GINsim. (SBML 199 kb)
Additional file 9: Stomatal opening levels for simulated single node
knockouts in the simplified model under all input signal combinations.
(XLSX 15 kb)
Acknowledgements
The authors thank Zhongyao Sun, Jorge G. T. Zañudo and Prof. Sarah
Assmann for helpful discussions.
Funding
This project was supported by National Science Foundation (NSF) grants IIS
1160995, PHY 1205840 and MCB 1244303. The NSF had no role in the design
of the study, analysis and interpretation of data or in writing the manuscript.
Availability of data and materials
The datasets supporting the conclusions of this article are included within
the article and its additional files.
Authors’ contributions
XG developed the reduced model and performed the analysis under the
advice and supervision of RA. Both authors wrote the manuscript. Both
authors have read and approved the final version of the manuscript.
Competing interests
The authors declare that they have no competing interests.
Consent for publication
Not applicable. The article does not contain any individual person’s data.
Ethics approval and consent to participate
Not applicable. The article does not involve human participants, human data
or human tissue.
Received: 10 June 2016 Accepted: 11 August 2016
References
1.
Stigler B, Chamberlin HM. A regulatory network modeled from wild-type
gene expression data guides functional predictions in Caenorhabditis
elegans development. BMC Syst Biol. 2012;6:77.
2.
Chifman J, et al. The core control system of intracellular iron homeostasis: a
mathematical model. J Theor Biol. 2012;300:91–9.
3.
Massague J. TGF-beta signal transduction. Annu Rev Biochem. 1998;67:753–91.
4.
Xu HL, et al. Construction and Validation of a Regulatory Network for
Pluripotency and Self-Renewal of Mouse Embryonic Stem Cells. PLoS
Comput Biol. 2014;10(8):e1003777.
5.
Kestler HA, et al. Network modeling of signal transduction: establishing the
global view. Bioessays. 2008;30(11–12):1110–25.
6.
Tyson JJ, Chen K, Novak B. Network dynamics and cell physiology. Nat Rev
Mol Cell Biol. 2001;2(12):908–16.
7.
Thomas R, European Molecular Biology Organization. Kinetic logic : a
Boolean approach to the analysis of complex regulatory systems :
proceedings of the EMBO course "Formal analysis of genetic regulation,"
held in Brussels, September 6–16, 1977, Lecture notes in biomathematics.
Berlin; New York: Springer; 1979. p. xiii, 507.
8.
Kauffman SA. Metabolic stability and epigenesis in randomly constructed
genetic nets. J Theor Biol. 1969;22(3):437.
9.
Glass L, Kauffman SA. Logical analysis of continuous, nonlinear biochemical
control networks. J Theor Biol. 1973;39(1):103–29.
10.
Wang RS, Saadatpour A, Albert R. Boolean modeling in systems biology: an
overview of methodology and applications. Phys Biol. 2012;9(5):055001.
11.
Miskov-Zivanov N, et al. The duration of T cell stimulation is a critical
determinant of cell fate and plasticity. Sci Signal. 2013;6(300):ra97.
12.
Chaouiya C, Naldi A, Thieffry D. Logical modelling of gene regulatory
networks with GINsim. Methods Mol Biol. 2012;804:463–79.
13.
Deritei D, et al. Principles of dynamical modularity in biological regulatory
networks. Sci Rep. 2016;6:21957.
14.
Murrugarra D, Laubenbacher R. Regulatory patterns in molecular interaction
networks. J Theor Biol. 2011;288:66–72.
15.
Sun Z, et al. Multi-level modeling of light-induced stomatal opening
offers new insights into its regulation by drought. PLoS Comput Biol.
2014;10(11):e1003930.
16.
Li S, Assmann SM, Albert R. Predicting essential components of signal
transduction networks: a dynamic model of guard cell abscisic acid
signaling. PLoS Biol. 2006;4(10):e312.
17.
Schroeder JI, et al. Guard Cell Signal Transduction. Annu Rev Plant Physiol
Plant Mol Biol. 2001;52:627–58.
18.
Shimazaki K, et al. Light regulation of stomatal movement. Annu Rev Plant
Biol. 2007;58:219–47.
19.
Assmann SM. Enhancement of the Stomatal Response to Blue Light by Red
Light, Reduced Intercellular Concentrations of CO(2), and Low Vapor
Pressure Differences. Plant Physiol. 1988;87(1):226–31.
20.
Bergmann DC, Sack FD. Stomatal development. Annu Rev Plant Biol.
2007;58:163–81.
Gan and Albert BMC Systems Biology (2016) 10:78
Page 13 of 14
21.
MacArthur BD, Ma'ayan A, Lemischka IR. Systems biology of stem cell fate
and cellular reprogramming. Nat Rev Mol Cell Biol. 2009;10(10):672–81.
22.
Steinway SN, et al. Network modeling of TGFbeta signaling in hepatocellular
carcinoma epithelial-to-mesenchymal transition reveals joint sonic hedgehog
and Wnt pathway activation. Cancer Res. 2014;74(21):5963–77.
23.
Saadatpour A, Albert R, Reluga TC. A Reduction Method for Boolean
Network Models Proven to Conserve Attractors. SIAM J Appl Dyn Syst. 2013;
12(4):1997–2011.
24.
Ansotegui C, Manya F. Mapping problems with finite-domain variables
to problems with Boolean variables. Theory Appl of Satisfiability Test.
2005;3542:1–15.
25.
Van Ham P. How to deal with more than two levels. In: Thomas R, editor.
Kinetic logic : a Boolean approach to the analysis of complex regulatory
systems : proceedings of the EMBO course "Formal analysis of genetic
regulation," held in Brussels, September 6–16, 1977. Berlin; New York:
Springer; 1979. p. 326–44.
26.
Didier G, Remy E, Chaouiya C. Mapping multivalued onto Boolean
dynamics. J Theor Biol. 2011;270(1):177–84.
27.
Karlsson PE. Blue light regulation of stomata in wheat seedlings. I. Influence
of red background illumination and initial conductance level. Physiol Plant.
1986;66:5.
28.
Zanudo JG, Albert R. An effective network reduction approach to find
the dynamical repertoire of discrete dynamic networks. Chaos. 2013;
23(2):025111.
29.
Chaouiya C, et al. SBML qualitative models: a model representation format
and infrastructure to foster interactions between qualitative modelling
formalisms and tools. BMC Syst Biol. 2013;7:135.
30.
Veliz-Cuba A, et al. Steady state analysis of Boolean molecular network
models via model reduction and computational algebra. BMC
Bioinformatics. 2014;15:221.
31.
Remy E, Ruet P, Thieffry D. Graphic requirements for multistability and
attractive cycles in a Boolean dynamical framework. Adv Appl Math. 2008;
41(3):335–50.
32.
Remy E, Ruet P. On differentiation and homeostatic behaviours of Boolean
dynamical systems. Lect Notes Bioinformatics. 2007;4780:92–101.
33.
Richard A, Comet J-P. Necessary conditions for multistationarity in discrete
dynamical systems. Discret Appl Math. 2007;155(18):2403–13.
34.
Richard A. Negative circuits and sustained oscillations in asynchronous
automata networks. Adv Appl Math. 2010;44(4):378–92.
35.
Naldi A, et al. Dynamically consistent reduction of logical regulatory graphs.
Theor Comput Sci. 2011;412(21):2207–18.
36.
Liu YY, Slotine JJ, Barabasi AL. Controllability of complex networks. Nature.
2011;473(7346):167–73.
37.
Lin CT. Structural controllability. IEEE Trans Autom Control. 1974;AC19(3):201–8.
38.
Mochizuki A, et al. Dynamics and control at feedback vertex sets. II: A
faithful monitor to determine the diversity of molecular activities in
regulatory networks. J Theor Biol. 2013;335:130–46.
39.
Zanudo JG, Albert R. Cell fate reprogramming by control of intracellular
network dynamics. PLoS Comput Biol. 2015;11(4):e1004193.
40.
Schwartz A, et al. Anion-Channel Blockers Inhibit S-Type Anion Channels
and Abscisic Acid Responses in Guard Cells. Plant Physiol. 1995;109(2):651–8.
41.
Kim TH, et al. Guard cell signal transduction network: advances in
understanding abscisic acid, CO2, and Ca2+ signaling. Annu Rev Plant Biol.
2010;61:561–91.
42.
Albert R, Othmer HG. The topology of the regulatory interactions predicts
the expression pattern of the segment polarity genes in Drosophila
melanogaster. J Theor Biol. 2003;223(1):1–18.
• We accept pre-submission inquiries
• Our selector tool helps you to find the most relevant journal
• We provide round the clock customer support
• Convenient online submission
• Thorough peer review
• Inclusion in PubMed and all major indexing services
• Maximum visibility for your research
Submit your manuscript at
www.biomedcentral.com/submit
Submit your next manuscript to BioMed Central
and we will help you at every step:
Gan and Albert BMC Systems Biology (2016) 10:78
Page 14 of 14
|
27542373
|
PP1cc_2 = ( ( ( PLD AND ( ( ( NOT phot1_complex AND NOT BL ) ) ) ) AND NOT ( PLD_high ) ) OR ( ( phot1_complex AND ( ( ( NOT PLD ) ) ) ) AND NOT ( PLD_high ) ) OR ( ( BL AND ( ( ( NOT PLD ) ) ) ) AND NOT ( PLD_high ) ) ) OR NOT ( PLD OR phot1_complex OR BL OR PLD_high )
Kv = ( Kc )
carbfix_high = ( Ci AND ( ( ( phph_high ) ) ) ) OR ( CO2 AND ( ( ( phph_high ) ) ) )
sucrose = ( ( PLD ) AND NOT ( ABA ) )
Kin = ( FFA AND ( ( ( NOT Ci_sup OR NOT Ci ) AND ( ( ( PMV_neg ) ) ) ) ) ) OR ( ABA AND ( ( ( NOT Ci_sup OR NOT Ci ) AND ( ( ( PMV_neg ) ) ) ) ) ) OR ( PMV_neg AND ( ( ( NOT Cac_high AND NOT ABA AND NOT Ci_sup AND NOT FFA ) ) ) )
PLD = ( ABA ) OR ( NO )
PLA2 = ( BL ) OR ( RL ) OR ( phot1_complex )
Kout = ( PMV_pos AND ( ( ( Ci ) AND ( ( ( Ci_sup ) ) ) ) OR ( ( NOT ROS ) ) OR ( ( NOT NO ) ) OR ( ( ABA ) ) OR ( ( NOT FFA ) ) ) )
Cac = ( ABA ) OR ( ( CaR ) AND NOT ( CaATPase ) ) OR ( ( CaIc ) AND NOT ( CaATPase ) )
PK_2 = ( ( ( Ci_sup AND ( ( ( PP1cc_1 ) AND ( ( ( NOT PP1cc_3 AND NOT PP1cc_2 ) ) ) ) OR ( ( PP1cc_3 AND PP1cc_2 ) AND ( ( ( NOT PP1cc_1 ) ) ) ) OR ( ( PP1cc_1 AND PP1cc_2 ) AND ( ( ( NOT PP1cc_3 ) ) ) ) ) ) AND NOT ( Ci ) ) OR ( PP1cc_1 AND ( ( ( Ci ) AND ( ( ( NOT Ci_sup AND NOT PP1cc_2 ) ) ) ) ) ) OR ( ( ( PP1cc_3 AND ( ( ( NOT PP1cc_1 AND NOT PP1cc_2 ) ) ) ) AND NOT ( Ci_sup ) ) AND NOT ( Ci ) ) OR ( ( ( PP1cc_2 AND ( ( ( NOT PP1cc_3 AND NOT PP1cc_1 ) ) OR ( ( PP1cc_3 AND PP1cc_1 ) ) OR ( ( PP1cc_1 ) AND ( ( ( NOT PP1cc_3 ) ) ) ) ) ) AND NOT ( Ci_sup ) ) AND NOT ( Ci ) ) ) OR NOT ( PP1cc_3 OR PP1cc_1 OR Ci_sup OR PP1cc_2 OR Ci )
FFA = ( PLA2 )
HATPase_3 = ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( FFA AND ( ( ( phph_high AND PK_2 AND PK_3 ) AND ( ( ( NOT PK_1 AND NOT PLA2 ) ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( phph_high ) ) AND NOT ( phph_high ) ) AND NOT ( phph_high ) ) OR ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( PLA2 AND ( ( ( PK_1 AND PK_3 ) AND ( ( ( NOT PK_2 AND NOT FFA ) ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( phph AND ( ( ( NOT phph_high ) ) ) ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( phph_high ) ) AND NOT ( phph_high ) ) AND NOT ( phph_high ) ) AND NOT ( phph_high ) ) AND NOT ( phph_high ) )
KEV = ( Cac_high AND ( ( ( Kv ) ) ) )
Kc = ( ( ( ( ( ( HATPase_3 AND ( ( ( HATPase_2 AND Kv AND KEV AND AnionCh ) ) OR ( ( HATPase_2 AND Kin AND AnionCh ) ) ) ) AND NOT ( Kout ) ) AND NOT ( Kout ) ) AND NOT ( AnionCh ) ) AND NOT ( AnionCh_high ) ) AND NOT ( AnionCh_high ) ) OR ( ( ( ( HATPase_2 AND ( ( ( Kin ) ) OR ( ( Kv AND KEV ) ) ) ) AND NOT ( Kout ) ) AND NOT ( AnionCh ) ) AND NOT ( AnionCh_high ) ) OR ( ( HATPase_1 AND ( ( ( Kin ) ) OR ( ( Kv ) AND ( ( ( KEV ) ) ) ) ) ) AND NOT ( Kout ) )
SO_2 = ( HATPase_2 AND ( ( ( Kv ) ) ) ) OR ( sucrose AND ( ( ( NOT Kv ) ) ) )
NO = ( phph AND ( ( ( ROS ) ) ) )
CaR = ( PLC ) OR ( NO )
PLD_high = ( ABA AND ( ( ( NO ) ) ) )
HATPase_2 = ( ( ( ( ( ( FFA AND ( ( ( phph_high AND PK_2 AND PK_1 ) AND ( ( ( NOT PK_3 ) ) ) ) ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) OR ( ( ( ( ( PLA2 AND ( ( ( PK_2 AND phph ) AND ( ( ( NOT PK_1 AND NOT FFA AND NOT PK_3 ) ) ) ) ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) ) AND NOT ( Cac_high ) )
ROS = ( phph AND ( ( ( PLD ) AND ( ( ( NOT ABI1 ) ) ) ) ) )
SO_3 = ( HATPase_3 AND ( ( ( Kv ) ) ) )
PMV_neg = ( ( ( HATPase_3 AND ( ( ( NOT PMV_pos ) ) OR ( ( AnionCh AND PMV_neg ) ) ) ) AND NOT ( Cac_high ) ) AND NOT ( KEV ) ) OR ( ( PMV_neg AND ( ( ( Cac_high ) AND ( ( ( HATPase_2 OR HATPase_3 OR HATPase_1 ) ) ) ) OR ( ( PMV_pos ) AND ( ( ( NOT Cac_high AND NOT HATPase_2 AND NOT KEV AND NOT HATPase_3 AND NOT AnionCh AND NOT HATPase_1 ) ) ) ) OR ( ( NOT Cac_high AND NOT HATPase_2 AND NOT KEV AND NOT PMV_pos AND NOT HATPase_3 AND NOT AnionCh AND NOT HATPase_1 ) ) OR ( ( NOT Cac_high ) AND ( ( ( HATPase_2 OR HATPase_3 OR HATPase_1 ) ) ) ) ) ) AND NOT ( AnionCh ) ) OR ( ( ( HATPase_2 AND ( ( ( NOT PMV_pos ) ) OR ( ( AnionCh AND PMV_neg ) ) ) ) AND NOT ( Cac_high ) ) AND NOT ( KEV ) ) OR ( ( ( HATPase_1 AND ( ( ( NOT PMV_pos ) ) OR ( ( AnionCh AND PMV_neg ) ) ) ) AND NOT ( Cac_high ) ) AND NOT ( KEV ) )
MCPS = ( Ci AND ( ( ( RL OR BL ) ) ) ) OR ( Ci_sup AND ( ( ( RL OR BL ) ) ) )
AnionCh_high = ( ( ABA ) AND NOT ( ABI1 ) ) OR ( Ci AND ( ( ( Ci_sup ) ) ) ) OR ( ( Cac_high ) AND NOT ( ABI1 ) )
SO_1 = ( HATPase_1 AND ( ( ( Kv ) ) ) )
phph = ( BL ) OR ( RL )
AnionCh = ( ( ( ( ABA AND ( ( ( ABI1 ) AND ( ( ( NOT phot1_complex AND NOT BL ) ) ) ) OR ( ( NOT Cac_high AND NOT ABI1 AND NOT Ci_sup AND NOT Ci ) ) OR ( ( NOT ABI1 ) AND ( ( ( Ci_sup ) ) OR ( ( Ci ) ) ) ) ) ) AND NOT ( ABI1 ) ) AND NOT ( ABI1 ) ) OR ( ( Cac_high ) AND NOT ( ABI1 ) ) OR ( Ci_sup AND ( ( ( NOT Cac_high AND NOT ABA AND NOT ABI1 AND NOT phot1_complex AND NOT BL ) ) ) ) OR ( Ci AND ( ( ( Ci_sup ) ) ) ) OR ( ABI1 AND ( ( ( NOT ABA AND NOT phot1_complex AND NOT BL ) ) ) ) ) OR NOT ( Cac_high OR ABA OR ABI1 OR phot1_complex OR Ci_sup OR Ci OR BL )
PLC = ( BL ) OR ( ABA AND ( ( ( Cac ) ) ) )
ABI1 = NOT ( ( ABA ) )
PP1cc_1 = ( BL ) OR ( phot1_complex )
Cac_high = ( ( CaR AND ( ( ( ABA ) ) ) ) AND NOT ( CaATPase ) ) OR ( ( CaIc AND ( ( ( ABA ) ) ) ) AND NOT ( CaATPase ) )
Ci = ( CO2 AND ( ( ( NOT MCPS_high AND NOT CO2_high AND NOT carbfix_high ) ) OR ( ( CO2_high ) ) ) )
phot1_complex = ( BL )
PK_1 = ( ( ( PP1cc_1 AND ( ( ( NOT PP1cc_2 ) ) OR ( ( PP1cc_2 ) AND ( ( ( NOT PP1cc_3 ) ) ) ) ) ) AND NOT ( Ci ) ) AND NOT ( Ci_sup ) ) OR ( ( Ci_sup AND ( ( ( PP1cc_1 AND PP1cc_2 ) AND ( ( ( NOT PP1cc_3 ) ) ) ) OR ( ( PP1cc_3 AND PP1cc_1 ) AND ( ( ( NOT PP1cc_2 ) ) ) ) ) ) AND NOT ( Ci ) ) OR ( ( Ci AND ( ( ( PP1cc_1 AND PP1cc_2 ) ) AND ( ( NOT PP1cc_3 ) ) ) ) AND NOT ( Ci_sup ) ) OR ( ( ( PP1cc_2 AND ( ( ( PP1cc_3 ) AND ( ( ( NOT PP1cc_1 ) ) ) ) ) ) AND NOT ( Ci ) ) AND NOT ( Ci_sup ) )
HATPase_1 = ( ( PLA2 AND ( ( ( PK_2 AND PK_1 AND PK_3 AND phph ) AND ( ( ( NOT FFA ) ) ) ) OR ( ( PK_2 AND PK_3 AND phph ) AND ( ( ( NOT PK_1 AND NOT FFA ) ) ) ) OR ( ( PK_1 AND phph ) AND ( ( ( NOT PK_2 AND NOT FFA AND NOT PK_3 ) ) ) ) OR ( ( PK_1 AND PK_3 ) AND ( ( ( NOT PK_2 AND NOT FFA ) ) ) ) OR ( ( PK_2 AND PK_1 ) AND ( ( ( NOT FFA ) ) ) ) ) ) AND NOT ( Cac_high ) ) OR ( ( FFA AND ( ( ( PK_2 AND PK_3 AND phph ) AND ( ( ( NOT PK_1 ) ) ) ) OR ( ( PK_1 AND phph ) AND ( ( ( NOT PK_2 ) ) ) ) OR ( ( PK_2 AND PK_1 ) AND ( ( ( NOT PK_3 ) ) ) ) OR ( ( PK_1 AND PK_3 ) AND ( ( ( NOT PK_2 ) ) ) ) OR ( ( PK_2 AND PK_1 AND PK_3 ) ) ) ) AND NOT ( Cac_high ) )
PP1cc_3 = ( ( PLD_high AND ( ( ( NOT phot1_complex AND NOT BL ) ) ) ) OR ( BL AND ( ( ( PLD ) AND ( ( ( NOT PLD_high ) ) ) ) ) ) OR ( phot1_complex AND ( ( ( PLD ) AND ( ( ( NOT PLD_high ) ) ) ) ) ) ) OR NOT ( PLD OR phot1_complex OR BL OR PLD_high )
PK_3 = ( PP1cc_1 AND ( ( ( PP1cc_2 ) AND ( ( ( NOT PP1cc_3 AND NOT Ci_sup AND NOT Ci ) ) ) ) OR ( ( PP1cc_3 AND Ci_sup ) AND ( ( ( NOT Ci AND NOT PP1cc_2 ) ) ) ) OR ( ( PP1cc_3 AND Ci ) AND ( ( ( NOT Ci_sup AND NOT PP1cc_2 ) ) ) ) ) )
MCPS_high = ( Ci AND ( ( ( RL AND BL ) ) ) ) OR ( Ci_sup AND ( ( ( RL AND BL ) ) ) )
CaIc = ( PMV_neg ) OR ( ROS )
phph_high = ( RL AND ( ( ( BL ) ) ) )
carbfix = ( Ci AND ( ( ( phph ) ) ) ) OR ( CO2 AND ( ( ( phph ) ) ) )
Ci_sup = ( CO2 AND ( ( ( MCPS_high OR CO2_high OR carbfix_high ) ) ) )
CaATPase = ( Cac )
PMV_pos = ( ( ( Cac_high AND ( ( ( NOT HATPase_2 AND NOT HATPase_3 AND NOT HATPase_1 ) ) ) ) AND NOT ( AnionCh ) ) AND NOT ( PMV_neg ) ) OR ( AnionCh AND ( ( ( NOT HATPase_2 AND NOT HATPase_3 AND NOT HATPase_1 ) AND ( ( ( Cac_high ) ) ) ) OR ( ( PMV_pos ) AND ( ( ( NOT Cac_high AND NOT HATPase_2 AND NOT HATPase_3 AND NOT HATPase_1 AND NOT PMV_neg ) ) ) ) OR ( ( KEV ) AND ( ( ( NOT Cac_high AND NOT HATPase_2 AND NOT HATPase_3 AND NOT HATPase_1 ) ) ) ) ) ) OR ( ( ( KEV AND ( ( ( NOT HATPase_2 AND NOT HATPase_3 AND NOT HATPase_1 ) ) ) ) AND NOT ( AnionCh ) ) AND NOT ( PMV_neg ) ) OR ( ( ( ( PMV_pos AND ( ( ( KEV AND AnionCh ) AND ( ( ( HATPase_2 OR HATPase_3 OR HATPase_1 ) ) ) ) OR ( ( Cac_high AND AnionCh ) AND ( ( ( HATPase_2 OR HATPase_3 OR HATPase_1 ) ) ) ) ) ) AND NOT ( AnionCh ) ) AND NOT ( PMV_neg ) ) AND NOT ( PMV_neg ) )
|
ORIGINAL RESEARCH
published: 19 August 2016
doi: 10.3389/fphys.2016.00349
Frontiers in Physiology | www.frontiersin.org
1
August 2016 | Volume 7 | Article 349
Edited by:
Christian Diener,
National Institute of Genomic
Medicine, Mexico
Reviewed by:
Oksana Sorokina,
University of Edinburgh, UK
Marcio Luis Acencio,
Norwegian University of Science and
Technology, Norway
*Correspondence:
Luis Mendoza
lmendoza@biomedicas.unam.mx
Rosana Pelayo
rosanapelayo@gmail.com
Specialty section:
This article was submitted to
Systems Biology,
a section of the journal
Frontiers in Physiology
Received: 22 March 2016
Accepted: 02 August 2016
Published: 19 August 2016
Citation:
Enciso J, Mayani H, Mendoza L and
Pelayo R (2016) Modeling the
Pro-inflammatory Tumor
Microenvironment in
Acute Lymphoblastic Leukemia
Predicts a Breakdown of
Hematopoietic-Mesenchymal
Communication Networks.
Front. Physiol. 7:349.
doi: 10.3389/fphys.2016.00349
Modeling the Pro-inflammatory
Tumor Microenvironment in
Acute Lymphoblastic Leukemia
Predicts a Breakdown of
Hematopoietic-Mesenchymal
Communication Networks
Jennifer Enciso 1, 2, Hector Mayani 1, Luis Mendoza 3* and Rosana Pelayo 1*
1 Oncology Research Unit, Mexican Institute for Social Security, Mexico City, Mexico, 2 Biochemistry Sciences Program,
Universidad Nacional Autónoma de Mexico, Mexico City, Mexico, 3 Departamento de Biología Molecular y Biotecnología,
Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
Lineage fate decisions of hematopoietic cells depend on intrinsic factors and
extrinsic signals provided by the bone marrow microenvironment, where they reside.
Abnormalities in composition and function of hematopoietic niches have been proposed
as key contributors of acute lymphoblastic leukemia (ALL) progression. Our previous
experimental findings strongly suggest that pro-inflammatory cues contribute to
mesenchymal niche abnormalities that result in maintenance of ALL precursor cells
at the expense of normal hematopoiesis. Here, we propose a molecular regulatory
network interconnecting the major communication pathways between hematopoietic
stem and progenitor cells (HSPCs) and mesenchymal stromal cells (MSCs) within the BM.
Dynamical analysis of the network as a Boolean model reveals two stationary states that
can be interpreted as the intercellular contact status. Furthermore, simulations describe
the molecular patterns observed during experimental proliferation and activation.
Importantly, our model predicts instability in the CXCR4/CXCL12 and VLA4/VCAM1
interactions following microenvironmental perturbation due by temporal signaling from
Toll like receptors (TLRs) ligation. Therefore, aberrant expression of NF-κB induced
by intrinsic or extrinsic factors may contribute to create a tumor microenvironment
where a negative feedback loop inhibiting CXCR4/CXCL12 and VLA4/VCAM1 cellular
communication axes allows for the maintenance of malignant cells.
Keywords: cancer systems biology, acute lymphoblastic leukemia, tumor microenvironment, CXCL12,
pro-inflammatory bone marrow, early hematopoiesis, network modeling, dynamical systems
INTRODUCTION
Cancer is currently considered as a global child health priority (Gupta et al., 2014). The
application of effective treatments to decrease overall childhood cancer mortality requires a
comprehensive understanding of its origins and pathobiology, along with accurate diagnosis and
early identification of high-risk groups (reviewed in Vilchis-Ordoñez et al., 2016). Strikingly, the
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
clinical, molecular and biological heterogeneity of malignant
diseases indicating an unsuspected multiclonal diversity has
highlighted their complexity and the uncertainty of their
cell population dynamics. Novel theoretical and experimental
integrative strategies have changed our perspective of cancer,
from a hierarchical, deterministic and unidirectional process to a
multi-factorial network where genetics interacts with micro and
macro environmental cues that contribute to the etiology and
maintenance of tumor cells (Notta et al., 2011; Davila-Velderrain
et al., 2015; Tomasetti and Vogelstein, 2015). Furthermore,
stochastic effects associated with the number of stem cell
divisions have been proposed as major contributors, often even
more significant than hereditary or external factors (Tomasetti
and Vogelstein, 2015).
B-cell acute lymphoblastic leukemia (B-ALL) is largely the
result of a growing number of cooperating genetic and epigenetic
aberrations that corrupt hematopoietic developmental pathways
and ultimate lead to uncontrolled production of malignant
B lymphoid precursor cells within the bone marrow (BM)
(Pelayo et al., 2012; Purizaca et al., 2012). Leukemic cell
infiltration and treatment failure worsen the outcome of the
disease and remain the foremost cause of relapse. Recent
advances suggest the ability of leukemia initiating cells to create
abnormal BM microenvironments, promoting high proliferation
and early differentiation arrest at the expense of normal cell
fate decisions (Colmone et al., 2008; Raaijmakers, 2011; Vilchis-
Ordoñez et al., 2015). Intrinsic damage and/or remodeling of cell
compartments that shape the distinct BM niches may account
to microenvironmental regulation of quiescence, proliferation,
differentiation and blastic cell migration. Leukemic cells compete
for niche resources with their normal hematopoietic counterparts
(Wu et al., 2009), culminating in the displacement of the
latter, as observed in xenotransplantation mice models (Colmone
et al., 2008). Moreover, the marrow microenvironment provides
leukemic precursors with dynamic interactions and regulatory
signals that are essential for their maintenance, proliferation
and survival. Although, the underlying molecular mechanisms
are poorly defined, these niches protect tumor cells from
chemotherapy-induced apoptosis, showing a new perspective on
the evolution of chemoresistance (Ayala et al., 2009: Shain et al.,
2015; Tabe and Konopleva, 2015), and emphasizing the need
for new models that theoretically or experimentally replicate the
interplay between tumor and stromal cells under normal and
pathological settings.
As suggested by our previous findings, ALL lymphoid
precursors have the ability of responding to pathogen- or
damage- associated molecular patterns via Toll-like receptor
signaling
by
secreting
soluble
factors
and
altering
their
differentiation potentials (Dorantes-Acosta et al., 2013). The
resulting pro-inflammatory microenvironment may expose them
to prolonged proliferation, contributing tumor maintenance
in a self-sustaining way while prompting the NF-κB-associated
proliferation
of
normal
progenitor
cells
(Vilchis-Ordoñez
et al., 2015, 2016). Some hematopoietic growth factors and
pro-inflammatory
cytokines,
including
granulocyte-colony
stimulating factor (G-CSF), IFNα, IL-1α, IL-1β, IL-7, and TNFα
were highly produced by ALL cells from a conspicuous group of
patients co-expressing myeloid markers (Vilchis-Ordoñez et al.,
2015). Of note, mesenchymal stromal cells (MSCs) from ALL
BM have shown atypical production of pro-inflammatory factors
whereas disruption of the major cell communication pathway
is apparent by detriment of CXCL12 expression and biological
function (Geay et al., 2005; Colmone et al., 2008; van den Berk
et al., 2014).
Considering that the CXCL12/CXCR4 axis constitutes the
most critical component of the perivascular and reticular BM
niches supporting the hematopoietic stem and progenitor cells
(HSPCs) differentiation and maintenance within the BM, as
well as the early steps of B cell development (Ma et al., 1998;
Tokoyoda et al., 2004; Sugiyama et al., 2006; Greenbaum et al.,
2013), an obstruction of the HSPC-MSC interaction may have
substantial implications in the overall stability of these processes.
Whether the inflammation-derived signals provide a mechanism
for leukemic cells to survive, to induce changes in lineage cell fate
decisions, or to prompt niche remodeling in leukemia settings,
are currently topical questions.
Mathematical
model
strategies
have
become
powerful
approaches to complex biological systems and may contribute
to
unravel
the
hematopoietic-microenvironment
interplay
that facilitates tumor cells prevalence (Altrock et al., 2015;
Enciso et al., 2015). Through continuous dynamic modeling
with differential equations we have learned seminal aspects
of multi-compartment and multi-clonal behavior of leukemic
cell populations (Stiehl and Marciniak-Czochra, 2012; Enciso
et al., 2015), leading to novel proposals on disease development
driven by unbalanced competition between normal and pre-
leukemic cells (Swaminathan et al., 2015). Both stochastic and
deterministic models have been useful to simulate cell fate
decisions and predict clonal evolution (reviewed in Enciso et al.,
2015). Certainly, incorporating tumor microenvironment in
cancer modeling is expected to change our vision of biochemical
interactions in niche remodeling-dependent hematopoietic
growth, as recently demonstrated for myeloma disease (Coelho
et al., 2016).
By
developing
and
simulating
a
dynamic
Boolean
system, we now investigate the biological consequences of
microenvironmental
perturbation
due
by
temporal
TLR
signaling
on
crucial
communication
networks
between
stem/progenitor cells (HSPCs) and MSCs in ALL. We propose
that NF-κB dependent tumor-associated inflammation co-
participate in malignant progression concomitant to normal
hematopoietic failure through disruption of CXCL12/CXCR4
and VLA4/VCAM-1 communication axes.
MATERIALS AND METHODS
Manual Curation Strategy
Based on the crucial and unique role of the CXCL12/CXCR4
axis in the regulation of maintenance, biological activity, and
niche communication-derived cell fate decisions of seminal cells,
including pluripotent embryonic stem cells and multipotent
hematopoietic stem cells, construction and updating of molecular
interactions of relevance involved careful manual curation
of primary hematopoietic cell research. Moreover, of special
Frontiers in Physiology | www.frontiersin.org
2
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
interest was the attention to the hematopoietic malignancies,
which in contrast to solid tumors, display a distinct CXCL12-
mediated microenvironmental behavior. Thus, although the
modeled signaling pathways could be considered generic to all
tissues, the organ, stage of cell differentiation and surrounding
microenvironment may influence the net result of interactions.
Taking into account this considerations, most published work
that has been used for the reconstruction of our proposed
model, include data from molecular interactions in HSPCs.
Some of the interactions have been reported in a number
of different tissues and predicted to be conserved in the
hematopoietic system. Finally, as there is not enough data
to model hematopoietic-microenvironment restricted to Homo
sapiens and some interactions might be crucial for the molecular
connectivity of the model, we have used information from
different species when needed. A detailed referencing of all
reports used for the model reconstruction is provided as
Supplemental Material (Tables S1, S2, and reference list).
Molecular Basis for the Network
Reconstruction
The
connectivity
among
key
molecules
involved
in
the
communication between HSPCs and MSCs within the BM
was inferred through the curated experimental literature.
Specifically, we were interested in recovering the network
components, their interactions, and the nature of the interactions
(activation/positive or inactivation/negative). The resulting
general network incorporates transcriptional factors, kinases,
membrane receptors, interleukines, integrins, growth factors, and
chemokines from Homo sapiens and Mus musculus species.
Importantly, to simplify the modeling process, some groups
of molecules were considered as single functional modules,
thus encompassing a series of sequential steps that lead to the
activation or inactivation of a certain node (e.g., PI3K/Akt).
The following paragraphs summarize the principle evidence
used to reconstruct the HSPC-MSC network and infer the
logical rules for computational simulation of the system as a
discrete dynamical model. A detailed referencing is provided as
Supplemental Material (Tables S1, S2, and reference list).
The CXCR4/CXCL12 chemokine pathway was considered as
the central axis for the network construction considering its
essential role in homeostasis maintenance (Sugiyama et al., 2006;
Tzeng et al., 2011) and B lineage support (Ma et al., 1998;
Tokoyoda et al., 2004). Furthermore, recent observations suggest
that this axis is disrupted by up-stream molecular deregulations
both in MSC and leukemic blasts harvested from ALL patients,
affecting the maintenance of hematopoietic cells within their
regulatory niches (Geay et al., 2005; Colmone et al., 2008; van
den Berk et al., 2014). Besides the well-studied CXCR4/CXCL12
chemotactic interaction, CXCR4 activation increases the affinity
between vascular cellular adhesion molecule-1 (VCAM-1)
expressed on the surface of MSC and its receptor VLA-4 on
HSPC. Both pathways, CXCR4/CXCL12 and VLA-4/VCAM-1,
are known to play coordinately a central role in HSPC migration,
engraftment and retention within the BM (Peled et al., 2000;
Ramirez et al., 2009), converge in triggering the PI3K/Akt and
ERK signals, and share common up-stream regulators involving
molecular factors guiding inflammatory responses.
As mentioned in the Introduction, recent evidence indicates
the secretion of high levels of pro-inflammatory cytokines
by a conspicuous group of ALL patients (Vilchis-Ordoñez
et al., 2015), thereby presumably contributing to remodeling of
the normal hematopoietic microenvironment (Colmone et al.,
2008). Of note, interleukin-1α (IL-1α) and IL-1β, which were
substantially elevated, play an amplification role on inflammation
increasing the expression of other cytokines, like G-CSF
(Majumdar et al., 2000; Allakhverdi et al., 2013), and setting
a positive feedback loop with the PI3K co-activation of NF-
κB (Reddy et al., 1997; Sizemore et al., 1999; Carrero et al.,
2012; Bektas et al., 2014). IL-1 and G-CSF, inhibit directly and
indirectly the CXCR4/CXCL12 axis. G-CSF negatively regulates
CXCL12 transcription and increases the secretion of matrix
metalloproteinase-9, showing the ability to degrade both CXCL12
(Lévesque et al., 2003; Semerad et al., 2005; Christopher et al.,
2009; Day et al., 2015) and CXCR4 (Lévesque et al., 2003).
Moreover, G-CSF promotes up-regulation of Gfi1 that at the
time inhibits the transcription of CXCR4 (Zhuang et al., 2006;
De La Luz Sierra et al., 2007; de la Luz Sierra et al., 2010).
Thus, by considering this information from experimental data,
we have included IL-1 and G-CSF as key elements of the BM
microenvironment in the HSPC-MSC communication network.
In concordance, we incorporated as a “positive control
condition” an input node representing the Toll-like receptor
ligand (lTLR) lipopolysaccharide (LPS), that binds TLR4
and
triggers
the
conventional
and
well-known
NF-κB-
dependent
pro-inflammatory
response,
promoting,
among
other transcriptional targets, the transcription of pro-IL-1β
(Jones et al., 2001; Tak and Firestein, 2001; Wang et al., 2002;
Khandanpour et al., 2010; Higashikuni et al., 2013).
Downstream NF-κB, the expression of CXCR7 has been
shown to be upregulated (Tarnowski et al., 2010), which in
turn, down-regulates CXCR4 by heterodimerization, promoting
its internalization and further degradation. In parallel, activated
CXCR7 presents a higher affinity for CXCL12 and β-arrestin,
reducing CXCR4 signaling in CXCR7 and CXCR4 expressing
cells (Uto-Konomi et al., 2013; Coggins et al., 2014). However,
CXCR7 is unable to couple with G-protein, transducing through
recruitment of β-arrestin and leading to MAP kinases Akt and
ERK activation (Tarnowski et al., 2010; Uto-Konomi et al.,
2013; Torossian et al., 2014). As with CXCR4, CXCR7, and
VLA-4 activation in HSPC, PI3K/Akt pathway is activated on
HSPC and MSC, via G-CSF receptor signaling (Liu et al., 2007;
Vagima et al., 2009; Ponte et al., 2012; Furmento et al., 2014),
and after LPS stimulation (Guha and Mackman, 2002; Wang
et al., 2009; McGuire et al., 2013). Apparently, PI3K/Akt acts
at overlapping levels on the modulation of inflammation. On
the one hand, it increases the production of IL-1 antagonist
molecules (Williams et al., 2004; Molnarfiet al., 2005; Li and
Smith, 2014) and inhibits secretion of mature IL-1β (Tapia-
Abellán et al., 2014). On the other hand, it promotes nuclear
translocation of the transcriptional factor Foxo3a (Brunet et al.,
1999; Miyamoto et al., 2008; Park et al., 2008), down-regulating
indirectly the transcription of antioxidant enzymes and enabling
Frontiers in Physiology | www.frontiersin.org
3
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
reactive oxygen species (ROS) accumulation, which in turn
promotes maturation of pro-IL-1β and IL-1β secretion (Hsu and
Wen, 2002; Yang et al., 2007; Gabelloni et al., 2013).
At the mesenchymal counterpart, in addition to a number
of molecules participating in the MSC-subsystem sensitivity
to
microenvironmental
cues,
we
incorporated
an
input
node representing Gap-junction conformed by connexin-43
(Cx43)
that
mediates
direct
intercellular
communication
between mesenchymal cells. Strikingly, its integral activity as
calcium channel conductor has been shown to be a potent
positive regulator of CXCL12 transcription and secretion
(Schajnovitz et al., 2011).
Furthermore, Cx43 expression
appears to be critically disregulated in the BM stromal
cells from acute leukemia patients, suggesting an important
role in the hypothetic disregulation of the hematopoietic-
stromal intercellular communication (Liu et al., 2010; Zhang
et al., 2012). The inclusion of GSK3β and β-catenin in both
subsystems was relevant due to their roles as intermediates
of
signaling
transduction
and
regulation
of
the
main
intracellular communication elements proposed for our network
reconstruction. The model is available in XML format (GINML)
on GINsim Model Repository page (http://ginsim.org/models_
repository) (Chaouiya et al., 2012), under the title “HSPCs-
MSCs. Communication pathways between Hematopoietic Stem
Progenitor Cells (HSPCs) and MSCs.”
Dynamical Modeling of the HSPC-MSC
Network
For the computational modeling of the HSPC-MSC complex
system, we followed the standard steps to convert it into a discrete
dynamical system, as described by Albert and Wang (2009) and
Assman and Albert (2009). The Boolean approach is useful when
quantitative and detailed kinetic information is lacking. In such
a case, each node of the network is represented as a binary
element, allowed only to have an “active” (ON) or “inactive”
(OFF) state, numerically represented by 1 and 0, respectively.
The activation state of each node is dependent on the activation
state of its regulators, as described by Boolean functions, also
called logical rules. The classical Boolean operators employed
in Boolean functions are AND (&), OR (|) and NOT (!).
The AND operator is used to represent the requirement of
the conjunction of two or more nodes participating in the
regulation of a certain node (e.g., VLA-4 = CXCR4 & VCAM-
1 representing that VLA-4 optimal activation requires its ligand
VCAM-1 and the signaling due to CXCR4 activation). When
there is more than one node able to regulate another, but only
one of them is sufficient to exert the effect, the OR operator
is applied (e.g., PI3K/Akt = GCSF | ROS | TLR representing
that the activation of the G-CSF receptor, the increase of
intracelular ROS concentration or the binding of a TLR ligand
may activate PI3K/Akt signaling). Finally, the NOT operator
represents repression of a node over another (e.g., IL-1 =
(NF-κB & ROS) & !PI3K/Akt meaning that IL-1 requires the
transcriptional activation of pro-IL-1 promoted by NF-κB and
the post-transcriptional maturation mediated by ROS, but its
signaling is inhibited by the presence of PI3K/Akt). Detailed
compiling of reviewed references for the network reconstruction
and the development of the logical rules can be found in Tables
S1, S2.
Given that each node in the network has an activation
state, then the general state of a network at a given time
t can be represented by a vector of n elements, where n
is the number of nodes in the network. For example, the
vector (00000010000000000100001000), represents a network
state where only the 7th, 18th, and 23rd elements are active.
In our model, this particular state represents the pattern of
activation where only GSK3B_H, GSK3B_M, and VCAM1_M are
active. Now, since we are implementing a dynamical system, it is
necessary to specify how the network may evolve from a time t to
t+1.
There are two possible implementations to model the
transition from one state of the network to another. On one
side, the synchronous scheme update the activation state of
all the nodes each time-step, assuming that all the biological
processes involved in the model occur at similar time scales.
And on the other side, asynchronous scheme update only one
of the logical rules per time step, considering a more complex
behavior of biological processes where molecular signaling is
likely to change at different time points depending on the
nature of the interaction (Albert and Wang, 2009). Either one
or another update scheme, take an initial combination of the
nodes (initial state) and update the logical rules successively
through an established number of time steps or until an steady
state or attractor is reached. Attractors may be of a single state
(fixed point attractors) or a set of states (cyclic or complex
attractors depending if they have one or more possible transition
paths among their constituent states). The analysis of the nodes
activation pattern in the attractors give the biological significance
of the computational simulations of the models (Albert and
Wang, 2009; Assman and Albert, 2009).
The dynamical behavior of the network was analyzed
implementing the logical rules into BoolNet (R open-source
package), and obtaining its attractors (stationary states) by
applying asynchronous update strategies (Müssel et al., 2010).
Under the asynchronous updating scheme, the simulation was
performed using 50,000 random initial states, updating the
network until either a fixed point attractor or a complex attractor
was reached. Confidence of the model was tested through
the simulation of all possible mutants (constitutive and null
activation of every node) and the comparison of the resultant
attractors with experimental reports about the biological effects
in vivo or in vitro after the use of antagonists or the generation of
knock-in and knock-out models.
Dynamical Multicellular Approach
Assuming that every simulation beginning at a certain initial
state of the network represents the dynamical profile of a
single cell, Wu and collaborators proposed a “population-like”
analysis for a discrete model (Wu et al., 2009). Similarly, we
asynchronously ran the simulations of the network from 50,000
random initial states, and then updated for 2000 time-steps,
followed by calculation of the average activation value from
50,000 simulations for each node in each time-step. Such data was
Frontiers in Physiology | www.frontiersin.org
4
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
plotted as multi-cellular average activation graphs. Furthermore,
we evaluated the effect of a short (1 time-step) and a sustained
(699 time-steps) temporary induction of lTLR in time-step 700
and 1400, and analyzed the dynamical effects in the wild type
network and in some relevant mutant networks.
RESULTS
Network Reconstruction
The
inferred
HSPC-MSC
network
(Figure 1)
constitutes
the
first
attempt
to
model
relevant
interaction
axes
between undifferentiated hematopoietic cells and the BM
microenvironment,
that
may
approach
us
to
a
deeper
understanding of the numerous molecular signals influencing
the
hematopoietic
system
regulation
during
normal
and
malignant processes. Our current ALL network has 26 nodes
and 80 interactions. Among them, twelve nodes correspond
to molecules that are expressed in HSPC and involved in
intracellular signaling (PI3K/Akt, Gfi1, NF-κB, GSK3β, FoxO3a,
ERK, β-catenin, and ROS) or cell-membrane receptors for
communication with the microenvironment (CXCR4, CXCR7,
VLA-4, and TLR). Eleven nodes conform the MSC subsystem,
integrated by intracellular signaling molecules (PI3K/Akt,
NF-κB, GSK3β, FoxO3a, ERK, β-catenin, and ROS), a gap-
junction protein regulating communication among MSC (Cx43),
communication ligands with HSPC (VCAM-1 and CXCL12)
and TLR. Common internal nodes in both HSPC and MSC
systems are representative molecules from the most studied
pathways influencing proliferation, migration, survival, and
-some of them- differentiation. Finally, the microenvironmental
compartment is represented by G-CSF secreted by myeloid
and stromal cells (Majumdar et al., 2000; Allakhverdi et al.,
2013; Tesio et al., 2013; Boettcher et al., 2014), its inductor
IL-1 which is secreted by MSC and HSPC, and lTLR so as
FIGURE 1 | Regulatory HSPC-MSC network. The network is constituted by three compartments represented with different geometric shapes: HSPC, MSC, and
microenvironmental soluble factors. HSPC and MSC have intracellular nodes regulating the response and expression of elements mediating the communication
between them. CXCR4-CXCL12 and VLA-4/VCAM-1 axes are suggested to be the most crucial communicating elements. HSPC and MSC are both susceptible of
TLR stimulation with lTLR input. HSPC, hematopoietic stem and progenitor cell; MSC, mesenchymal stromal cell.
Frontiers in Physiology | www.frontiersin.org
5
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
TABLE 1 | Logical rules used for HSPC-MSC modeling as a Boolean
system on BoolNet.
Node
Logical rule
Bcatenin_H
!GSK3B_H
CXCR4_H
CXCL12_M & !(CXCR7_H | GCSF | Gfi1_H)
CXCR7_H
CXCL12_M & NfkB_H
ERK_H
((CXCR4_H & PI3KAkt_H) | CXCR7_H | GCSF | Gfi1_H | ROS_H |
VLA4_H ) & !(FoxO3a_H | GSK3B_H)
FoxO3a_H
(Bcatenin_H | ROS_H) & !(ERK_H | PI3KAkt_H)
Gfi1_H
(GCSF | TLR_H) & !Gfi1_H
GSK3B_H
!PI3KAkt_H
NfkB_H
(TLR_H | ROS_H | (IL1 & PI3KAkt_H)) &
!(FoxO3a_H)
PI3KAkt_H
((CXCR4_H & CXCR7_H) | GCSF | ROS_H | TLR_H | VLA4_H) &
!FoxO3a_H
ROS_H
IL1 & TLR_H & (!FoxO3a_H)
TLR_H
lTLR
VLA4_H
VCAM1_M & CXCR4_H
Cx43_M
Cx43_M
Bcatenin_M
!(FoxO3a_M | GSK3B_M | NfkB_M)
CXCL12_M
Cx43_M & !(Bcatenin_M | GCSF | NfkB_M)
ERK_M
GCSF | ROS_M | TLR_M
FoxO3a_M
(Bcatenin_M | ROS_M) & !(ERK_M | PI3KAkt_M)
GSK3B_M
!PI3KAkt_M
NfkB_M
(IL1 & PI3KAkt_M) | (ROS_M & ERK_M) | TLR_M
ROS_M
IL1 & TLR_M & (!FoxO3a_M)
PI3KAkt_M
GCSF | ROS_M | TLR_M
TLR_M
lTLR
VCAM1_M
!Bcatenin_M | NfkB_M | PI3KAkt_M
lTLR
lTLR
IL1
((ROS_M | NfkB_M) & !PI3KAkt_M) | ((ROS_H | NfkB_H) &
!PI3KAkt_H)
GCSF
IL1
Nodes representing molecules in HSPC are denoted with “_H” at the end of the node
name, while nodes representing molecules in MSC are denoted with “_M.” Logical rules
were constructed using the logical operators AND ( & ), OR ( | ) and NOT ( ! ). The
corresponding common names and genes ID are found in Table S3.
to model a homeostasis disruption that is known to drive a
pro-inflammatory signaling. Model inputs are Cx43 and lTLR,
while the activation value of the other 24 nodes is dependent on
the network topology and the initial state of the input nodes. All
logical rules used for the computational simulation with BoolNet
are shown in Table 1. Note that the logical rules for the input
nodes include self-regulations, but these are for computational
purposes to represent their sustained activation, rather than a
biological reality.
Attractors of the Wild-Type Network:
Searching for the Relevance of TLR in the
Biology of CXCL12
The asynchronous simulation of the Boolean model returned
4
attractors:
2
fixed
points
and
2
complex
attractors
(Figure 2). The first two attractors, fixed point attractor
1 and 2, were identified with the physiological detached
and attached state of the HSPC with its MSC counterpart,
respectively.
Both fixed point attractors will depend on the initial states of
both, TLR and Cx43. Thus, in the absence of lTLR, the final fates
will depend on the initial activation state of Cx43. However, once
TLR is activated, final fates are not contributed anymore from the
activation state of Cx43.
Loss of HSPC-MSC communication corresponding to a
detachment state, is due to the absence of Cx43 and the
consequent inactivation of CXCL12. In the activation pattern of
this attractor, only VCAM-1 accompanied by GSK3β in both sub
systems remained active (Tabe et al., 2007). On the contrary,
when Cx43 is active (as in fixed point attractor 2), CXL12
is expressed by the MSC, which in turn positively regulates
the CXCR4 receptor required for the activation of the VLA-
4/VCAM-1 axis. The pattern in HSPC, correspond to ERK
and PI3K/Akt activation, well-described elements downstream
CXCR4 and VLA-4 (Tabe et al., 2007). β-catenin, a subject of
debate about its function on stem cell maintenance, is turned
on as a consequence of the GSK3β inhibition by PI3K/Akt (Dao
et al., 2007).
Complex attractors 1 and 2 share the same activation values
in all nodes, except for the initial state of Cx43 which is an
input and therefore may be consistently either active or inactive
through simulation. Importantly, these two attractors have the
node for ITLR active, so that under induced pro-inflammatory
conditions the resultant perturbation of CXCR4/CXCL12 and
VLA-4/VCAM-1 is exclusively dependent on CXCL12 down
regulation in MSC by NF-κB. The network attractors are
concordant with experimental observations (Ueda et al., 2004;
Wang et al., 2012; Yi et al., 2012) with the exception of IL1
and GCSF inactivation although lTLR-induced NF-κB signaling
in hematopoietic and mesenchymal compartments. In order
to explain this discrepancy we may remark that an attractor
is a stable network state or set of states, reached after the
network went through a sequence of transient states where, in
most biological systems, there is cross-pathway communication
for modulating cellular response (Williams et al., 2004; Tapia-
Abellán et al., 2014), so IL1 and GCSF could be activated in
some transient states but down-regulated by other pathways
responding to lTLR activation. Due to the existence of regulatory
circuits among pathways, in the presence of ITLR there is an
oscillatory behavior of ERK and Gfi1. Therefore, we applied
the dynamic multicellular approach described by Wu et al.
(2009) in order to have a deeper understanding of the HSPC-
MSC model upon perturbations. The average activation value of
50,000 simulations for all nodes within the HSPC-MSC network
was plotted and presented in Figure 3. The plots represent a
qualitative approach for the analysis of the cell population trend
under specific conditions. Considering that the initial activation
values are randomly chosen, with exception of lTLR, TLR_M,
and TLR_H which activation value was set to 0, the average
initial activation value for the rest of the nodes correspond to 0.5.
From time-step 0 to time-step 499 correspond to the stabilization
of the dynamics. Of note, the plateau obtained around time-
steps 500-699 corresponds to the average of the two fixed point
attractors.
Frontiers in Physiology | www.frontiersin.org
6
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
FIGURE 2 | Asynchronous attractors from the wild type network. Dark color denotes an activation value of 1, while light color represents an activation value =
0. The blue, orange, and yellow colors distinguish the nodes in the three compartments in the HSPC-MSC network corresponding to HSPC, MSC, and
microenvironmental factors, respectively. The last two attractors obtained when the initial states for the asynchronous simulation had lTLR value = 1, have two nodes
(ERK_H and Gfi1_H) whose activation values oscillate and are responsible of the complex attractor. Oscillatory values are represented by intermediate blue color.
Nodes representing molecules in HSPC are denoted with “_H” at the end of the node name, while nodes representing molecules in MSC are denoted with “_M.”
Analysis of Transitory States Applied to a
Multicellular Approach: from
Pro-inflammatory Signals to CXCL12
Downregulation
The short lTLR stimulation at time-step 700 and 1400
(Figures 3A–C)
induces
up-regulation
of
Gfi1
in
HSPC
(Figure 3A), and of NF-κB and PI3K/Akt in both HSPC and MSC
compartments (Figures 3A,B). These nodes maintain a sustained
activation as long as the lTLR is present (Figures 3D–F). In
contrast, ERK, ROS and FoxO3a showed an increase but are
regulated by other nodes, providing a feedback to basal values.
Accompanying the cross-regulation of intracellular pathways, a
decrease on CXCR4, CXCL12, VLA-4, and VCAM-1 activation
is observed. As expected, there is positive signaling of the pro-
inflammatory cytokines with a parallel co-increase of CXCR7,
signals damped by PI3K/Akt and CXCL12 down-regulation,
respectively.
Model Validation by Mutant Analysis
Listed in Table 2 are the observations from comparisons
between the resultant attractors of simulations with null
(“loss of function”) and constitutive expression mutants (“gain
of function”), against the wild-type model. We focused on
the activation value changes in the two axes of interest –
CXCR4/CXCL12 and VCAM-1/VLA-4. Even though the nodes
included in the reconstruction of the present model are
well-studied elements of cell fate related-pathways, there is
a lack of experiments correlating their perturbation with
microenvironment modifications that impact HSPC behavior
(Table 2, Table S4). Due to this missing data, and in order to
validate the model, we now used available information of general
alterations in hematopoiesis in the presence of lTLR.
MSC ERK, FoxO3a, and PI3K/Akt nodes participating in
CXCR4/CXCL12 and VCAM-1 VLA-4 axes regulation were not
found in the revised literature. β-catenin in MSC has a role
on osteoblastogenesis and its constitutive induced expression in
osteoblasts in a mice model results in acute myeloid leukemia
(AML) induction (Kode et al., 2014). The constitutive expression
of β-catenin showed an outcome where, under non-induced
inflammation, the CXCR4/CXCL12 axis is disrupted. This gives
support to our hypothesis that CXCR4/CXCL12 is probably
involved in the maintenance of leukemic cells. Furthermore, the
dynamic multicellular approach in the gain of function of β-
catenin in MSC, reproduced the recovery of VCAM-1 expression
upon stimulation of lTLR as reported by Kincade in OP9 cells
(Figure S1; Malhotra and Kincade, 2009).
GSK3β inhibition in MSC has been known to function in the
regulation of osteoblast and adipocyte differentiation. Besides,
experimental effect of a GSK3β-inhibitor on osteoblastogenesis
has shown that the decrease of this kinase induces down-
regulation of CXCL12 expression (Satija et al., 2013). The model
is consistent with the unsteadiness of CXCL12 activation in the
simulation of the mutant (Figures S2A,B).
According to our hypothesis, a pro-inflammatory-induced
CXCR4/CXCL12 disruption results in leukemic progression
support. In the proposed model, overexpression of NF-κB
disrupts the HSPC-MSC communication (Figure S2C). This is
in agreement with the reported leukocytosis associated to up-
regulation of NF-κB within BM MSCs from a mice model of
high-fat diet (Cortez et al., 2013). Finally, modeling of a gain of
function mutation in ROS resulted in the blocking of CXCL12
activation (Figure S2D). This is also in accordance of the recent
report of oxidative damage induced by iron in MSC, resulting
in down-regulation of CXCL12 expression and reduction of
their hematopoietic supporting function (Zhang et al., 2015).
Moreover, the iron-induced hematopoietic alterations previously
observed by other groups, are attenuated by the treatment with
ROS inhibitors (Lu et al., 2013).
Nodes in HSPC which have been experimentally reported
as dispensable for hematopoiesis, which did not show any
alterations in the CXCR4/CXCL12 and VLA-4/VCAM-1 axes on
the mutant simulations, are β-catenin (Figures S3A–D; Cobas
et al., 2004; Jeannet et al., 2008) and CXCR7 (Figures S3E,F).
Frontiers in Physiology | www.frontiersin.org
7
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
FIGURE 3 | Average activation value for intracellular HSPC nodes (A,D), intracellular MSC nodes (B,E) and communication axes among HSPC, MSC,
and microenvironment (C,F). (A–C) Correspond to simulations with a short (1 time-step) stimulation of lTLR at time-steps 700 and 1400. (D–F) Correspond to
simulations with lTLR stimulation at time-step 700 with a length of 699 time-steps. Nodes representing molecules in HSPC are denoted with “_H” at the end of the
node name, and nodes representing molecules in MSC are denoted with “_M.” Gray area covers the stabilization time steps until attractors are reached.
However, even though in vivo β-catenin null mutant HSPC does
not lose long-term reconstitution capacity or multipotentiallity,
its overexpression produces lose of stemness and differentiation
blockage to erythroid and lymphoid lineages (Kirstetter et al.,
2006; Scheller et al., 2006). Simulations of the gain of function
of β-catenin resulted in the appearance of additional attractors
where FoxO3a and GSK3β are increased (Figures S4A,B,
S5B), suggesting a reduction in proliferation and/or apoptosis
induction (Maurer et al., 2006; Yamazaki et al., 2006). In turn,
the simulation of overexpression of FoxO3a showed a down-
regulation of ERK and PI3K (Figures S4C, S5C). Also reported
as proliferative repressors in HSPC (Hock et al., 2004; Zeng
et al., 2004; Holmes et al., 2008), Gfi1 and GSK3β overexpression
mutants inhibited ERK activation, and additionally Gfi1 induce
the downregulation of PI3K/Akt node, CXCR4/CXCL12 and
VLA-4/VCAM-1 axes (Figures S4 and S5). Disagreeing with
experimental data (Holmes et al., 2008), GSK3β null mutant
outcome result in an additional attractor where PI3K/Akt and
ERK are inactive, notwithstanding CXCR4 and VLA4 activation
(Figure S6).
Of interest, NF-κB (Figure 4) and ROS (Figures S4F,
S5F)
constitutive
expression
in
HSPC
induce
additional
attractors with activation of IL-1 and G-CSF, and inhibition
of
axes
regulating
HSPC-MSC
contact.
A
number
of
investigations on cancer cells report a correlation of NF-
κB increased levels and CXCR4 (Richmond, 2002; Ayala
et
al.,
2009;
Shin
et
al.,
2014).
Nonetheless,
a
recent
study in human leukemic cell lines has shown that LPS
treatment
increases
MMP-9
activity,
a
metalloproteinase
known
to
efficiently
degrade
CXCR4
and
CXCL12
(Hajighasemi and Gheini, 2015).
NF-κB Gain of Function Mutant as ALL
Simplified Model
How common alterations in ALL cells may induce BM
microenvironment remodeling, regardless of the underlying
Frontiers in Physiology | www.frontiersin.org
8
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
TABLE 2 | Results from the model outcome for single node mutations.
Loss of function
Node
Model outcome
Experimental evidence
Bcatenin_H, CXCR7_H, ERK_H, FoxO3a_H, NfkB_H,
ROS_H, Bcatenin_M, ERK_M, FoxO3a_M, NfkB_M,
ROS_M, IL1, GCSF
No changes in the CXCR4/CXCL12 and VLA-4/VCAM-1 axes
with respect to the attractors from the wild-type model.
Cobas et al., 2004; Jeannet et al., 2008; Sierro
et al., 2007
CXCL12_M, CXCR4_H
Loss of CXCR4/CXCL12 and VLA-4/VCAM-1 in the fixed point
attractor with active Cx43_M.
Greenbaum et al., 2013; Sugiyama et al., 2006;
Tzeng et al., 2011
Gfi1_H
No changes in the CXCR4/CXCL12 and VLA-4/VCAM-1 axes.
Stabilization of lTLR-dependent complex attractors with no
activation of ERK_H .
Hock et al., 2004; Zeng et al., 2004
GSK3B_H
Additional fixed point attractor when Cx43 is active, where
FoxO3a_H is up-regulated and repressing PI3K_H and ERK_H.
Also, are additional complex attractor in the presence of lTLR
where FoxO3a_H inhibits PI3KAkt_H, ERK_H and NfkB_H
activation.
Holmes et al., 2008
PI3KAkt_H, PI3KAkt_M
No changes in CXCR4/CXCL12 and VLA-4/VCAM-1 axes with
respect to the attractors from the wild-type model. Under lTLR
stimulation, pro-inflammatory cytokines turned on and in
consequence ROS_H. In PI3KAkt_H null mutant, ERK_H is
inhibited in every condition and FoxO3a_H is intermittently
activated under lTLR stimulation.
Williams et al., 2004; Champelovier et al.,
2008; Xu et al., 2012
VLA-4, VCAM1_M
PI3KAkt_H and ERK_H are turned off even if CXCR4/CXCL12
axis is active.
Wang et al., 1998; Scott et al., 2003
GSK3B_M
Fixed point attractors are lost and became complex attractors.
Activation of Cx43, leads to two complex attractors of which one
activates CXCR4/CXCL12 and VLA-4/VCAM-1 axes
intermittently. In the absence of Cx43, two complex attractors are
generated, and one of them unsteadily activate IL1 and GCSF.
Satija et al., 2013
Gain of function
Node
Model outcome
GSK3B_M, ERK_M, VCAM1_M, FoxO3a_M
No changes in the CXCR4/CXCL12 and VLA-4/VCAM-1 axes
with respect to the attractors from the wild-type model.
NE (Not experimental evidence found)
CXCR7_H, NfkB_H, Bcatenin_M, NfkB_M, PI3KAkt_M,
GCSF, IL1
Loss of CXCR4/CXCL12 and VLA-4/VCAM-1 in the fixed point
attractor with active Cx43_M.
Cortez et al., 2013; Kode et al., 2014
Bcatenin_H
Under the activation of Cx43_M, an alternative steady state is
reached where PI3KAkt_H and ERK_H are not expressed and
instead, FoxO3a_H and GSK3B_H are active despite the
activation of CXCR4_H and VLA4_H.
Kirstetter et al., 2006; Champelovier et al.,
2008
CXCL12_M
Under lTLR stimulation, the complex attractors show a sustained
activation of CXCR7_H.
NE
FoxO3a_H
Bcatenin_H, ERK_H and PI3KAkt_H inactivation under any
condition.
Yamazaki et al., 2006
Gfi1_H
Loss of CXCR4/CXCL12 and VLA-4/VCAM-1 in the fixed point
attractor with active Cx43_M. Stabilization of lTLR-dependent
complex attractors.
Hock et al., 2004; Khandanpour et al., 2013
GSK3B_H
Inhibition of ERK_H and Bcatenin_H when CXCR4_H or lTLR are
active.
NE
PI3KAkt_H
Bcatenin_H remains active in the absence of Cx43 and lTLR.
Wang et al., 2013
ROS_H, ROS_M
Loss of CXCR4/CXCL12 and VLA-4/VCAM-1 in the fixed point
attractor with active Cx43_M. ROS_M overexpression mutant,
activates PI3K_M, which in consequence inhibits FoxO3a_M.
Lu et al., 2013; Zhang et al., 2015
VLA-4
Constitutive activation of PI3KAkt_H, ERK_H and Bcatenin_H.
Schofield et al., 1998; Shalapour et al., 2011
genetic aberration, was investigated by running a dynamic
multicellular simulation using the mutant network for NF-
κB gain of function within the HSPC sub-system. The results
shown in Figure 4 confirm that NF-κB mutation in HSPC
may perturb HSPC-MSC communication in parallel with
the induction of other alterations previously reported in
Frontiers in Physiology | www.frontiersin.org
9
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
FIGURE 4 | Dynamic multicellular simulation for a ALL simplified model
addressed by NF-κB gain of function in HSPC. Average activation for
intracellular HSPC nodes (A), intracellular MSC nodes (B) and communication
axes among HSPC, MSC, and microenvironment (C). Nodes representing
molecules in HSPC are denoted with “_H” at the end of the node name, while
nodes representing molecules in MSC are denoted with “_M.” Gray area
covers the stabilization time steps until attractors are reached.
ALL cells, such as the increase of Gfi1 expression (Purizaca
et al., 2013) and a pro-inflammatory milieu (Vilchis-Ordoñez
et al., 2015). IL1 and G-CSF activation by HSPC up-
regulate ERK, NF-κB and PI3K/Akt in MSC. As consequence
of PI3K/Akt increase in MSC, β-catenin is up-regulated
through the inhibition of GSK3β. Strikingly, the sustained
activation of CXCR7 resulted as a consequence of NF-
κB constitutive expression in HSPC and CXCL12 residual
expression from MSC. CXCR7/CXCL12 axis was recently
reported to be increased in ALL cells and a possible participation
in abnormal cell migration was suggested (Melo et al.,
2014).
DISCUSSION
According to the classical model of hematopoiesis, normal
blood cells are replenished throughout life by stem and early
progenitor populations undergoing stepwise differentiation
processes in the context of intersinusoidal specialized niches
(Purizaca et al., 2012; Vadillo et al., 2013). Cell cycle status,
self-renewing capability and the central cell fate decisions
depend, in great part, on the microanatomic organization and
signals from the BM environment. Endosteal, perivascular and
reticular niches provide support by cell-cell interactions and
growth/differentiation factors that control the expression of
lineage-specific transcription factors, among other elements.
Within the reticular niche, mainly composed by CXCL12-
abundant reticular cells (CARs), a special category of MSCs,
the chemokine CXCL12 and its receptor CXCR4 play a pivotal
role in the regulation of lymphopoiesis from the earliest
stages of the pathway (Tokoyoda et al., 2004; Nagasawa,
2015).
The
transcription
factor
Foxc1
governs
CXCL12
and stem cell factor expression, allowing the CAR niche
formation
for
maintenance
of
HSC,
common
lymphoid
progenitors, B cells, NK and plasmacytoid dendritic cells
(Omatsu et al., 2014). The net balance of its disruption is
instability of adaptive and innate immune cell production.
Recent findings suggest that elevation of cytokines and growth
factors, including G-CSF and TNFα, due to infectious stress,
substantially reduce the expression of CXCL12, SCF and
VCAM-1, further impairing primitive cell maintenance and
prompting their proliferation and migration (Kobayashi et al.,
2015, 2016).
Much remains to be unraveled about CXCL12-related
mechanisms
of
intercommunication
damage
that
may
favor growth of cancer cells at the expense of healthy
hematopoiesis
during
biological
contingencies
such
as
hematological malignancies and biological stress. Although,
genetic heterogeneity may be co-responsible for differences in
ALL overall survival, response to treatment, differentiation-stage
arrest or even predisposition to metastasis, a common need
might be the development of biological features that provide
pre-malignant cells decisive advantage over normal cells to
compete for the same ecological niche. Given the importance
of
CXCR4/CXCL12
axis
for
homeostatic
hematopoiesis
and of its presumptive disruption in ALL BM, we now
propose a Boolean model reconstructed with some of the
most studied elements upstream and downstream this key
communication axis. Our model shows its capacity to simulate
several phenotypes relevant to ALL. According to previous
experimental research, the major assumption made from
this model is that the integrity of CXCR4/CXCL12 signaling,
promoting the required activation of the VLA-4/VCAM-1
integrins interaction, is absolutely necessary for HSPC retention
in the mesenchymal niche and in consequence, indispensable
Frontiers in Physiology | www.frontiersin.org
10
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
for optimal hematopoiesis regulation (Lévesque et al., 2003;
Lua et al., 2012; Greenbaum et al., 2013; Park et al., 2013).
The
HSPC-MSC
model
asynchronous
simulation
in
the
absence of lTLR returned two attractors corresponding to
HSPC attachment and detachment to MSC. The ‘attachment’
status,
represented
by
the
induction
of
CXCR4/CXCL12
and/or VLA-4/VCAM-1 axes, also exhibited PI3K/Akt and
β-catenin activation within the HSPC compartment. Although
there is some controversy about the β-catenin role in HSC
regulation (Kirstetter et al., 2006; Duinhouwer et al., 2015),
the co-activation of PI3K/Akt and β-catenin is known to
promote
self-renewal
and
HSC
expansion
(Perry
et
al.,
2011).
Two
core
pathways
downstream
CXCR4/CXCL12
binding are PI3K/Akt and ERK, both promoters of cell
survival and regulators of proliferation. Considering that
the mesenchymal stromal niche has being identified as the
interface between the quiescence promoting osteoblastic niche
and the vascular niche regulating final lineage commitment
and cell migration, the signals provided by mesenchymal
cells
should
tightly
regulate
proliferation/expansion
in
order to further allow differentiation. According to this
statement,
the
attractor
representing
the
detached
state
conducts to pro-apoptosis signaling in the absence of aberrant
expression of NF-κB, that relies on cytochrome C release-
associated normal functions of GSK3β in HSPC (Maurer et al.,
2006).
By using elegant mice disease models and controlled culture
systems, a wealth body of studies has recently highlighted the co-
participation of inflammation and infectious stress in the HSPC
exit from quiescence status, as well as in cancer etiology and
progression (Baldridge et al., 2011; Vilchis-Ordoñez et al., 2015).
Chronic inflammation and carcinogenesis have been closely
connected via either a oncogenes-derived intrinsic pathway or
through an extrinsic pathway from external factors that promote
latent inflammatory responses involving signaling pathways such
as MyD88, NF-κB, and STAT3 (Mantovani et al., 2008; Krawczyk
et al., 2014).
Interestingly, pattern recognition receptors (PRRs), including
Toll-like receptors (TLRs) are functionally expressed from
the most primitive stages of hematopoiesis and contribute to
emergent cell replenishment in response to life-threatening
infections or disease-associated cell damage (Nagai et al., 2006;
Welner et al., 2008; Dorantes-Acosta et al., 2013; Vadillo et al.,
2014). This phenomenon is called emergency hematopoiesis and
is regulated at the most primitive cell level (Kobayashi et al., 2015,
2016).
The potential relevance of this mechanism in leukemogenesis
was the focus of this investigation, and our model allowed for the
analysis of most behaviors observed under experimental settings.
The discrete simulation of NF-κB constitutive expression mutant
on HSPC, gave further support to our hypothesis on the
perturbation of CXCR4/CXCL12 communication axis induced
by pro-inflammatory microenvironment. The single mutation
of NF-κB was sufficient to remodel the dynamical behavior of
the three sub-systems represented, which was an unexpected
behavior of the model. The dynamic analysis of the ALL-
like network, also suggested the activation of an alternative
communication pathway mediated by CXCR7 binding CXCL12.
Inhibition of CXCL12 within the mesenchymal niche, may
be fundamental for cell migration to adjacent BM structures
unable to sustain proper differentiation or even to extramedullar
tissues, accounting for a predictable role of this axis in
metastasis.
CONCLUDING REMARKS
The
proposed
HSPC-MSC
model
is
the
first
systemic
approximation
to
understand
the
intercommunication
pathways
underlying
primitive
cell
retention/proliferation
in
the mesenchymal niche as a
determinant factor
for
progression
of
hematological
hyperproliferative
diseases.
We applied conventional discrete dynamical modeling and
non-conventional population-like approaches as an average
behavior of the network model. Future improvement of discrete
dynamical modeling for ALL system will provide a powerful tool
for investigation of unbalanced competitions between leukemic
and normal hematopoietic cells within the BM. Overall, systems
biology will advance our comprehensive view of the mechanisms
involved in the pathogenesis of leukemic niches that may
illuminate therapeutic strategies based on cell-to-cell crosstalk
manipulation.
AUTHOR CONTRIBUTIONS
JE designed the work; generated, analyzed and interpreted
data; wrote the paper. HM interpreted data; revised the
work for intellectual content; wrote the paper. LM designed
the work; interpreted data; revised the work for intellectual
content; wrote the paper. RP designed the work; interpreted
data; revised the work for intellectual content; wrote the
paper.
ACKNOWLEDGMENTS
This work was supported by the National Council of Science
and Technology (CONACyT) (Grant CB-2010-01-152695 to RP),
by the Mexican Institute for Social Security (IMSS) (Grant
FIS/IMSS/PROT/G14/1289 to RP) and by the “Red Temática de
Células Troncales y Medicina Regenerativa” from CONACyT.
LM acknowledges the sabbatical scholarships from PASPA-DAPA
UNAM and CONACyT 251420. JE is scholarship holder from
CONACyT and IMSS, and was awarded by the PRODESI IMSS
Program.
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found
online
at:
http://journal.frontiersin.org/article/10.3389/fphys.
2016.00349
Frontiers in Physiology | www.frontiersin.org
11
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
REFERENCES
Albert, R., and Wang, R. S. (2009). Discrete dynamic modeling of cellular signaling
networks. Methods Enzymol. 467, 281–306. doi: 10.1016/S0076-6879(09)
67011-7
Allakhverdi, Z., Comeau, M. R., Armant, M., Agrawal, R., Woodfolk, J. A, Sehmi,
R., et al. (2013). Mast cell-activated bone marrow mesenchymal stromal cells
regulate proliferation and lineage commitment of CD34+ progenitor cells.
Front. Immunol. 4:461. doi: 10.3389/fimmu.2013.00461
Altrock, P. M., Liu, L. L., and Michor, F. (2015). The mathematics of cancer :
integrating quantitative models. Nat. Publ. Gr. 15, 730–745. doi: 10.1038/
nrc4029
Assman, S. M., and Albert, R. (2009). Discrete dynamic modelling with
asynchronous update or, how to model complex systems in the absence of
quantitative information. Methods Mol. Biol. 553, 207–225. doi: 10.1007/1-
4020-0613-6_16551
Ayala, F., Dewar, R., Kieran, M., and Kalluri, R. (2009). Contribution of bone
microenvironment to leukemogenesis and leukemia progression. Leukemia 23,
2233–2241. doi: 10.1038/leu.2009.175
Baldridge, M. T., King, K. Y., and Goodell, M. A. (2011). Inflammatory
signals regulate hematopoietic stem cells. Trends Immunol. 32, 57–65. doi:
10.1016/j.it.2010.12.003.Inflammatory
Bektas, A., Zhang, Y., Lehmann, E., Wood. W. H. III , Becker, K. G., Ferrucci, L.,
et al. (2014). Age-associated changes in basal NF-κB function in human CD4+
T lymphocytes via dysregulation of PI3 kinase. Aging (Albany. NY). 6, 957–974.
doi: 10.18632/aging.100705
Boettcher, S., Gerosa, R. C., Radpour, R., Bauer, J., Ampenberger, F., Heikenwalder,
M., et al. (2014). Endothelial cells translate pathogen signals into G-CSF-driven
emergency granulopoiesis. Blood 124, 1393–1403. doi: 10.1182/blood-2014-04-
570762
Brunet, A., Bonni, A., Zigmond, M. J., Lin, M. Z., Juo, P., Hu, L. S., et al. (1999).
Akt promotes cell survival by phosphorylating and inhibiting a forkhead
transcription factor. Cell 96, 857–868.
Carrero, R., Cerrada, I., Lled,ó, E., Dopazo, J., García-García, F., Rubio, M.-P.,
et al. (2012). IL1β induces mesenchymal stem cells migration and leucocyte
chemotaxis through NF-κB. Stem Cell Rev. 8, 905–916. doi: 10.1007/s12015-
012-9364-9
Champelovier, P., El Atifi, M., Pautre, V., Rostaing, B., Berger, F., and Seigneurin,
D. (2008). Specific inhibition of basal mitogen-activated protein kinases
and phosphatidylinositol 3 kinase activities in leukemia cells: a possible
therapeutic role for the kinase inhibitors. Exp. Hematol. 36, 28–36. doi:
10.1016/j.exphem.2007.08.027
Chaouiya, C., Naldi, A., and Thieffry, D. (2012). Logical modelling of gene
regulatory networks with GINsim. Methods Mol. Biol. 804, 463–479. doi:
10.1007/978-1-61779-361-5_23
Christopher, M. J., Liu, F., Hilton, M. J., Long, F., and Link, D. C. (2009).
Suppression of CXCL12 production by bone marrow osteoblasts is a common
and critical pathway for cytokine-induced mobilization. Blood 114, 1331–1339.
doi: 10.1182/blood-2008-10-184754
Cobas, M., Wilson, A., Ernst, B., Mancini, S. J. C., MacDonald, H. R., Kemler, R.,
et al. (2004). β-catenin is dispensable for hematopoiesis and lymphopoiesis. J.
Exp. Med. 199, 221–229. doi: 10.1084/jem.20031615
Coelho, R. M., Lemos, J. M., Alho, I., Valério, D., Ferreira, A. R., Costa, L., et al.
(2016). Dynamic modeling of bone metastasis, microenvironment and therapy:
integrating parathyroid hormone (PTH) effect, anti-resorptive and anti-cancer
therapy. J. Theor. Biol. 391, 1–12. doi: 10.1016/j.jtbi.2015.11.024
Coggins, N. L., Trakimas, D., Chang, S. L., Ehrlich, A., Ray, P., Luker, K.
E., et al. (2014). CXCR7 controls competition for recruitment of β-arrestin
2 in cells expressing both CXCR4 and CXCR7. PLoS ONE 9:e98328. doi:
10.1371/journal.pone.0098328
Colmone, A., Amorim, M., Pontier, A. L., Wang, S., Jablonski, E., and Sipkins,
D. A. (2008). Leukemic cells create bone marrow niches that disrupt the
behavior of normal hematopoietic progenitor cells. Science 322, 1861–1865. doi:
10.1126/science.1164390
Cortez, M., Carmo, L. S., Rogero, M. M., Borelli, P., and Fock, R. A. (2013). A
high-fat diet increases IL-1, IL-6, and TNF-α production by increasing NF-κb
and attenuating PPAR-γ expression in bone marrow mesenchymal stem cells.
Inflammation 36, 379–386. doi: 10.1007/s10753-012-9557-z
Dao, M. A., Creer, M. H., Nolta, J. A., and Verfaillie, C. M. (2007). Biology of
umbilical cord blood progenitors in bone marrow niches. Blood 110, 74–81.
doi: 10.1182/blood-2006-08-034447
Davila-Velderrain, J., Martinez-Garcia, J. C., and Alvarez-Buylla, E. R. (2015).
Modeling the epigenetic attractors landscape: toward a post-genomic
mechanistic understanding of development. Front. Genet. 6:160. doi: 10.3389/
fgene.2015.00160
Day, R. B., Bhattacharya, D., Nagasawa, T., and Link, D. C. (2015). Granulocyte
colony-stimulating factor reprograms bone marrow stromal cells to actively
suppress B lymphopoiesis in mice. Blood. doi: 10.1182/blood-2015-02-629444
De La Luz Sierra, M., Gasperini, P., McCormick, P. J., Zhu, J., and Tosato, G.
(2007). Transcription factor Gfi-1 induced by G-CSF is a negative regulator of
CXCR4 in myeloid cells. Blood 110, 2276–2285. doi: 10.1182/blood-2007-03-
081448
de la Luz Sierra, M., Sakakibara, S., Gasperini, P., Salvucci, O., Jiang, K.,
McCormick, P. J., et al. (2010). The transcription factor Gfi1 regulates G-CSF
signaling and neutrophil development through the Ras activator RasGRP1.
Blood 115, 3970–3979. doi: 10.1182/blood-2009-10-246967
Delgado-Martín, C., Escribano, C., Pablos, J. L., Riol-Blanco, L., and Rodríguez-
Fernández, J. L. (2011). Chemokine CXCL12 uses CXCR4 and a signaling core
formed by bifunctional akt, Extracellular Signal-regulated Kinase (ERK)1/2,
and Mammalian Target of Rapamycin Complex 1 (mTORC1) proteins to
control chemotaxis and survival simultaneously in mature dendriti. J. Biol.
Chem. 286, 37222–37236. doi: 10.1074/jbc.M111.294116
Doan, L. L., Porter, S. D., Duan, Z., Flubacher, M. M., Montoya, D., Tsichlis, P. N.,
et al. (2004). Targeted transcriptional repression of Gfi1 by GFI1 and GFI1B in
lymphoid cells. Nucleic Acids Res. 32, 2508–2519. doi: 10.1093/nar/gkh570
Dorantes-Acosta, E., Vadillo, E., Contreras-Quiroz, A., Balandrán, J. C., Arriaga-
Pizano, L., Purizaca, J., et al. (2013). TLR stimulation of bone marrow lymphoid
precursors from childhood Acute Leukemia modifies their differentiation
potentials. Biomed Res. Int. 2013:846724. doi: 10.1155/2013/846724
Duinhouwer, L. E., Tüysüz, N., Rombouts, E. W. J. C., Ter Borg, M. N. D.,
Mastrobattista, E., Spanholtz, J., et al. (2015). Wnt3a protein reduces growth
factor-driven expansion of human hematopoietic stem and progenitor cells
in serum-free cultures. PLoS ONE 10:e0119086. doi: 10.1371/journal.pone.
0119086
Enciso, J., Mendoza, L., and Pelayo, R. (2015). Normal vs. Malignant
hematopoiesis: the complexity of acute leukemia through systems biology.
Front. Genet. 6:290. doi: 10.3389/fgene.2015.00290
Essers, M. A. G., de Vries-Smits, L. M. M., Barker, N., Polderman, P. E., Burgering,
B. M. T., and Korswagen, H. C. (2005). Functional interaction between
β-catenin and FOXO in oxidative stress signaling. Science 308, 1181–1184. doi:
10.1126/science.1109083
Finn, N. A., and Kemp, M. L. (2012). Pro-oxidant and antioxidant effects of N-
acetylcysteine regulate doxorubicin-induced NF-kappa B activity in leukemic
cells. Mol. Biosyst. 8, 650–662. doi: 10.1039/c1mb05315a
Furmento, V. A., Marino, J., Blank, V. C., and Roguin, L. P. (2014). The
granulocyte colony-stimulating factor (G-CSF) upregulates metalloproteinase-
2 and VEGF through PI3K/Akt and Erk1/2 activation in human trophoblast
Swan
71
cells.
Placenta
35,
937–946.
doi:
10.1016/j.placenta.2014.
09.003
Gabelloni, M. L., Sabbione, F., Jancic, C., Bass, J. F., Keitelman, I., Iula, L.,
et al. (2013). NADPH oxidase derived reactive oxygen species are involved in
human neutrophil IL-1β secretion but not in inflammasome activation. Eur. J.
Immunol. 43, 3324–3335. doi: 10.1002/eji.201243089
Geay, J.-F., Buet, D., Zhang, Y., Foudi, A., Jarrier, P., Berthebaud, M., et al.
(2005). p210BCR-ABL inhibits SDF-1 chemotactic response via alteration of
CXCR4 signaling and down-regulation of CXCR4 expression. Cancer Res. 65,
2676–2683. doi: 10.1158/0008-5472.CAN-04-2152
Gorbunov, N. V., Garrison, B. R., McDaniel, D. P., Zhai, M., Liao, P. J.,
Nurmemet, D., et al. (2013). Adaptive redox response of mesenchymal stromal
cells to stimulation with lipopolysaccharide inflammagen: mechanisms of
remodeling of tissue barriers in sepsis. Oxid. Med. Cell. Longev. 2013:186795.
doi: 10.1155/2013/186795
Greenbaum, A., Hsu, Y.-M. S., Day, R. B., Schuettpelz, L. G., Christopher, M. J.,
Borgerding, J. N., et al. (2013). CXCL12 in early mesenchymal progenitors is
required for haematopoietic stem-cell maintenance. Nature 495, 227–230. doi:
10.1038/nature11926
Frontiers in Physiology | www.frontiersin.org
12
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
Guha, M., and Mackman, N. (2002). The phosphatidylinositol 3-kinase-Akt
pathway limits lipopolysaccharide activation of signaling pathways and
expression of inflammatory mediators in human monocytic cells. J. Biol. Chem.
277, 32124–32132. doi: 10.1074/jbc.M203298200
Gupta, S., Rivera-Luna, R., Ribeiro, R. C., and Howard, S. C. (2014). Pediatric
oncology as the next global child health priority: the need for national
childhood cancer strategies in low- and middle-income countries. PLoS Med.
11:e1001656. doi: 10.1371/journal.pmed.1001656
Hajighasemi, F., and Gheini, M. H. (2015). Lipopolysaccharide effect on vascular
endothelial factor and matrix metalloproteinases in leukemic cell lines In vitro.
Iran. J. Cancer Prev. 8:e2327. doi: 10.17795/ijcp2327
Higashikuni, Y., Tanaka, K., Kato, M., Nureki, O., Hirata, Y., Nagai, R.,
et al. (2013). Toll-like receptor-2 mediates adaptive cardiac hypertrophy in
response to pressure overload through interleukin-1β upregulation via nuclear
factor κB activation. J. Am. Heart Assoc. 2:e000267. doi: 10.1161/JAHA.113.
000267
Hock, H., Hamblen, M. J., Rooke, H. M., Schindler, J. W., Saleque, S., Fujiwara,
Y., et al. (2004). Gfi-1 restricts proliferation and preserves functional integrity
of haematopoietic stem cells. Nature 431, 1002–1007. doi: 10.1038/nature
02994
Holmes, T., O’Brien, T. A., Knight, R., Lindeman, R., Shen, S., Song, E., et al.
(2008). Glycogen synthase kinase-3β inhibition preserves hematopoietic stem
cell activity and inhibits leukemic cell growth. Stem Cells 26, 1288–1297. doi:
10.1634/stemcells.2007-0600
Hsu, H.-Y., and Wen, M.-H. (2002). Lipopolysaccharide-mediated reactive
oxygen species and signal transduction in the regulation of interleukin-1
gene expression. J. Biol. Chem. 277, 22131–22139. doi: 10.1074/jbc.M1118
83200
Jeannet, G., Scheller, M., Scarpellino, L., Duboux, S., Gardiol, N., Back, J., et al.
(2008). Long-term, multilineage hematopoiesis occurs in the combined absence
of β-catenin and γ-catenin. Blood 111, 142–149. doi: 10.1182/blood-2007-07-
102558
Jones, B. W., Means, T. K., Heldwein, K. A., Keen, M. A., Hill, P. J., Belisle, J. T.,
et al. (2001). Different Toll-like receptor agonists induce distinct macrophage
responses. J. Leukoc. Biol. 69, 1036–1044.
Khandanpour, C., Phelan, J. D., Vassen, L., Schütte, J., Chen, R., Horman, S. R.,
et al. (2013). Growth factor independence-1 antagonizes a p53-induced DNA
damage response pathway in lymphoblastic leukemia. Cancer Cell 23, 200–214.
doi: 10.1016/j.ccr.2013.01.011
Khandanpour, C., Sharif-askari, E., Vassen, L., Gaudreau, M. C., Zhu, J.,
Paul, W. E., et al. (2010). Evidence that Growth factor independence 1b
regulates dormancy and peripheral blood mobilization of hematopoietic
stem cells Evidence that Growth factor independence 1b regulates dormancy
and peripheral blood mobilization of hematopoietic stem cells. Blood 116,
5149–5161. doi: 10.1182/blood-2010-04-280305
Kirstetter, P., Anderson, K., Porse, B. T., Jacobsen, S. E. W., and Nerlov, C.
(2006). Activation of the canonical Wnt pathway leads to loss of hematopoietic
stem cell repopulation and multilineage differentiation block. Nat. Immunol. 7,
1048–1056. doi: 10.1038/ni1381
Kobayashi, H., Kobayashi, C. I., Nakamura-Ishizu, A., Karigane, D., Haeno, H.,
Yamamoto, K. N., et al. (2015). Bacterial c-di-GMP affects hematopoietic
Stem/progenitors and their niches through STING. Cell Rep. 11, 71–84. doi:
10.1016/j.celrep.2015.02.066
Kobayashi,
H.,
Suda,
T.,
and
Takubo,
K.
(2016).
How
hematopoietic
stem/progenitors and their niche sense and respond to infectious stress.
Exp. Hematol. 44, 92–100. doi: 10.1016/j.exphem.2015.11.008
Kode, A., Manavalan, J. S., Mosialou, I., Bhagat, G., Rathinam, C. V, Luo, N.,
et al. (2014). Leukemogenesis induced by an activating β-catenin mutation in
osteoblasts. Nature 506, 240–244. doi: 10.1038/nature11130
Krawczyk, J., O’Dwyer, M., Swords, R., Freeman, C., and Giles, F. J. (2014).
“The role of inflammation in leukaemia,” in Inflammation and Cancer, Vol.
816, Advances in Experimental Medicine and Biology, eds B. B. Aggarwal, B.
Sung, and S. C. Gupta (Basel: Springer), 335–360. doi: 10.1007/978-3-0348-
0837-8_13
Lévesque, J., Hendy, J., Takamatsu, Y., Simmons, P. J., and Bendall, L. J.
(2003). Disruption of the CXCR4 / CXCL12 chemotactic interaction during
hematopoietic stem cell mobilization induced by GCSF or cyclophosphamide.
J. Clin. Invest. 111, 187–196. doi: 10.1172/JCI200315994
Li, B., and Smith, T. J. (2014). PI3K/AKT pathway mediates induction of IL-1RA
by TSH in fibrocytes: modulation by PTEN. J. Clin. Endocrinol. Metab. 99,
3363–3372. doi: 10.1210/jc.2014-1257
Liu, H., Mihara, K., and Song, G. (2007). Interferon- γ attenuates the survival
activity of G-CSF through PI3K / Akt signaling pathway in mouse multipotent
progenitor cells. Ann. Hematol. 86, 547–555. doi: 10.1007/s00277-007-
0308-4
Liu, Y., Zhang, X., Li, Z., and Chen, X. (2010). Up-regulation of Cx43 expression
and GJIC function in acute leukemia bone marrow stromal cells post-
chemotherapy. Leuk. Res. 34, 631–640. doi: 10.1016/j.leukres.2009.10.013
Lu, W., Zhao, M., Rajbhandary, S., Xie, F., Chai, X., Mu, J., et al. (2013). Free iron
catalyzes oxidative damage to hematopoietic cells/mesenchymal stem cells in
vitro and suppresses hematopoiesis in iron overload patients. Eur. J. Haematol.
91, 249–261. doi: 10.1111/ejh.12159
Lua, J., Suna, Y., Nombela-Arrietaa, C., Dua, K. P., Parka, S.-Y., Chaib, L., et al.
(2012). Fak depletion in both hematopoietic and non-hematopoietic niche cells
leads to hematopoietic stem cell expansion. Exp. Hematol. 40, 307–317. doi:
10.1016/j.biotechadv.2011.08.010
Ma, Q., Jones, D., Borghesani, P. R., Segal, R. A., Nagasawa, T., Kishimoto, T.,
et al. (1998). Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar
neuron migration in CXCR4- and SDF-1-deficient mice. Proc. Natl. Acad. Sci.
U.S.A. 95, 9448–9453.
Majumdar, M. K., Thiede, M. A., Haynesworth, S. E., Bruder, S. P., and
Gerson, S. L. (2000). Human marrow-derived mesenchymal stem cells (MSCs)
express hematopoietic cytokines and support long-term hematopoiesis when
differentiated toward stromal and osteogenic lineages. J. Hematother. Stem Cell
Res. 9, 841–848. doi: 10.1089/152581600750062264
Malhotra, S., and Kincade, P. W. (2009). Canonical Wnt pathway signaling
suppresses VCAM-1 expression by marrow stromal and hematopoietic cells.
Exp. Hematol. 37, 19–30. doi: 10.1016/j.exphem.2008.08.008
Mantovani, A., Allavena, P., Sica, A., and Balkwill, F. (2008). Cancer-related
inflammation. Nature 454, 444. doi: 10.1038/nature07205
Maurer, U., Charvet, C., Wagman, A. S., Dejardin, E., and Green, D. R.
(2006). Glycogen synthase kinase-3 regulates mitochondrial outer membrane
permeabilization and apoptosis by destabilization of MCL-1. Mol. Cell 21,
749–760. doi: 10.1016/j.molcel.2006.02.009
McGuire, V. A., Gray, A., Monk, C. E., Santos, S. G., Lee, K., Aubareda, A.,
et al. (2013). Cross talk between the Akt and p38α pathways in macrophages
downstream of Toll-like receptor signaling. Mol. Cell. Biol. 33, 4152–4165. doi:
10.1128/MCB.01691-12
Melo, R. C. C., Longhini, A. L., Louzao Bigarella, C., Ozello Baratti, M., Traina,
F., Favaro, P., et al. (2014). CXCR7 is highly expressed in acute lymphoblastic
leukemia and potentiates CXCR4 response to CXCL12. PLoS ONE 9:e85926.
doi: 10.1371/journal.pone.0085926
Miyamoto, K., Miyamoto, T., Kato, R., Yoshimura, A., Motoyama, N., and Suda,
T. (2008). FoxO3a regulates hematopoietic homeostasis through a negative
feedback pathway in conditions of stress or aging. Blood 112, 4485–4493. doi:
10.1182/blood-2008-05-159848
Molnarfi, N., Hyka-Nouspikel, N., Gruaz, L., Dayer, J.-M., and Burger, D.
(2005). The production of IL-1 receptor antagonist in IFN- β-stimulated
human monocytes depends on the activation of Phosphatidylinositol 3-kinase
but not of STAT1. J. Immunol. 174, 2974–2980. doi: 10.4049/jimmunol.174.
5.2974
Müssel, C., Hopfensitz, M., and Kestler, H. A. (2010). BoolNet-an R package for
generation, reconstruction and analysis of Boolean networks. Bioinformatics 26,
1378–1380. doi: 10.1093/bioinformatics/btq124
Nagai, Y., Garrett, K. P., Ohta, S., Bahrun, U., Kouro, T., Akira, S., et al. (2006).
Toll-like receptors on hematopoietic progenitor cells stimulate innate immune
system replenishment. Immunity 24, 801–812. doi: 10.1016/j.immuni.2006.
04.008
Nagasawa, T. (2015). CXCL12/SDF-1 and CXCR4. Front. Immunol. 6:301. doi:
10.3389/fimmu.2015.00301
Notta, F., Mullighan, C. G., Wang, J. C., Poeppl, A., Doulatov, S., Phillips, L.
A., et al. (2011). Evolution of human BCR-ABL1 lymphoblastic leukaemia-
initiating cells. Nature 469, 362–367. doi: 10.1038/nature09733
Omatsu, Y., Seike, M., Sugiyama, T., Kume, T., and Nagasawa, T. (2014). Foxc1
is a critical regulator of haematopoietic stem/progenitor cell niche formation.
Nature 508, 536–540. doi: 10.1038/nature13071
Frontiers in Physiology | www.frontiersin.org
13
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
Park, S., Guo, J., Kim, D., and Cheng, J. Q. (2008). Identification of 24p3
as a direct target of Foxo3a regulated by Interleukin-3 through the
phosphoinositide 3-Kinase/Akt pathway. J. Biol. Chem. 284, 2187–2193. doi:
10.1074/jbc.M806131200
Park, S.-Y., Wolfram, P., Canty, K., Harley, B., Nombela-Arrieta, C., Pivarnik, G.,
et al. (2013). Focal adhesion kinase regulates the localization and retention of
pro-B cells in bone marrow microenvironments. J. Immunol. 190, 1094–1102.
doi: 10.4049/jimmunol.1202639
Pelayo, R., Dorantes-Acosta, E., Vadillo, E., and Fuentes-Panana, E. (2012). “From
HSC to B-lymphoid cells in normal and malignant hematopoiesis,” in Advances
in Hematopoietic Stem Cell Research, ed R. Pelayo (InTech), 277–298. doi:
10.5772/32213
Peled, A., Kollet, O., Ponomaryov, T., Petit, I., Franitza, S., Grabovsky, V., et al.
(2000). The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5
on immature human CD34(+) cells: role in transendothelial/stromal migration
and engraftment of NOD/SCID mice. Blood 95, 3289–3296.
Perry, J. M., He, X. C., Sugimura, R., Grindley, J. C., Haug, J. S., Ding, S.,
et al. (2011). Cooperation between both Wnt/β-catenin and PTEN/PI3K/Akt
signaling promotes primitive hematopoietic stem cell self-renewal and
expansion. Genes Dev. 25, 1928–1942. doi: 10.1101/gad.17421911
Ponte, A. L., Ribeiro-Fleury, T., Chabot, V., Gouilleux, F., Langonné, A., Hérault,
O., et al. (2012). Granulocyte-colony-stimulating factor stimulation of bone
marrow mesenchymal stromal cells promotes CD34+ Cell migration via
a matrix Metalloproteinase-2-Dependent mechanism. Stem Cells Dev. 21,
3162–3172. doi: 10.1089/scd.2012.0048
Purizaca, J., Contreras-Quiroz, A., Dorantes-Acosta, E., Vadillo, E., Arriaga-
Pizano, L., Fuentes-Figueroa, S., et al. (2013). Lymphoid progenitor cells
from childhood acute lymphoblastic leukemia are functionally deficient and
express high levels of the transcriptional repressor gfi-1. Clin. Dev. Immunol.
2013:349067. doi: 10.1155/2013/349067
Purizaca, J., Meza, I., and Pelayo, R. (2012). Early lymphoid development and
microenvironmental cues in B-cell acute lymphoblastic leukemia. Arch. Med.
Res. 43, 89–101. doi: 10.1016/j.arcmed.2012.03.005
Raaijmakers, M. H. G. P. (2011). Niche contributions to oncogenesis: emerging
concepts and implications for the hematopoietic system. Haematologica 96,
1041–1048. doi: 10.3324/haematol.2010.028035
Ramirez, P., Rettig, M. P., Uy, G. L., Deych, E., Holt, M. S., Ritchey, J. K., et al.
(2009). BIO5192, a small molecule inhibitor of VLA-4, mobilizes hematopoietic
stem and progenitor cells. Blood 114, 1340–1343. doi: 10.1182/blood-2008-10-
184721
Reddy, S. A., G., Huang, J. H., and Liao, W. S.-L. (1997). Phosphatidylinositol 3-
Kinase in Interleukin 1 Signaling: PHYSICAL INTERACTION WITH THE
INTERLEUKIN 1 RECEPTOR AND REQUIREMENT IN NF B AND AP-
1 ACTIVATION. J. Biol. Chem. 272, 29167–29173. doi: 10.1074/jbc.272.46.
29167
Richmond, A. (2002). NF-κB, chemokine gene transcription and tumour growth.
Nat. Rev. Immunol. 2, 664–674. doi: 10.1038/nri887
Satija, N. K., Sharma, D., Afrin, F., Tripathi, R. P., and Gangenahalli, G.
(2013). High throughput transcriptome profiling of lithium stimulated human
mesenchymal stem cells reveals priming towards Osteoblastic lineage. PLoS
ONE 8:55769. doi: 10.1371/journal.pone.0055769
Schajnovitz, A., Itkin, T., D’Uva, G., Kalinkovich, A., Golan, K., Ludin, A., et al.
(2011). CXCL12 secretion by bone marrow stromal cells is dependent on cell
contact and mediated by connexin-43 and connexin-45 gap junctions. Nat.
Immunol. 12, 391–398. doi: 10.1038/ni.2017
Scheller, M., Huelsken, J., Rosenbauer, F., Taketo, M. M., Birchmeier, W.,
Tenen, D. G., et al. (2006). Hematopoietic stem cell and multilineage defects
generated by constitutive β-catenin activation. Nat. Immunol. 7, 1037–1047.
doi: 10.1038/ni1387
Schofield, K. P., Humphries, M. J., de Wynter, E., Testa, N., and Gallagher, J. (1998).
The Effect of α4β1-Integrin binding sequences of fibronectin on growth of cells
from human hematopoietic progenitors. Blood 91, 3230–3238.
Scott, L. M., Priestley, G. V, and Papayannopoulou, T. (2003). Deletion of
α4 integrins from adult hematopoietic cells reveals roles in homeostasis,
regeneration,
and
homing.
Mol.
Cell.
Biol.
23,
9349–9360.
doi:
10.1128/MCB.23.24.9349-9360.2003
Semerad, C. L., Christopher, M. J., Liu, F., Short, B., Simmons, P. J., Winkler, I.,
et al. (2005). G-CSF potently inhibits osteoblast activity and CXCL12 mRNA
expression in the bone marrow. Blood 106, 3020–3027. doi: 10.1182/blood-
2004-01-0272
Shain, K. H., Dalton, W. S., and Tao, J. (2015). The tumor microenvironment
shapes hallmarks of mature B-cell malignancies. Oncogene 34, 1–10. doi:
10.1038/onc.2014.403
Shalapour, S., Hof, J., Kirschner-Schwabe, R., Bastian, L., Eckert, C., Prada, J.,
et al. (2011). High VLA-4 expression is associated with adverse outcome
and distinct gene expression changes in childhood B-cell precursor acute
lymphoblastic leukemia at first relapse. Haematologica 96, 1627–1635. doi:
10.3324/haematol.2011.047993
Shin, H. C., Seo, J., Kang, B. W., Moon, J. H., Chae, Y. S., and Lee, S. J.
(2014). Clinical significance of nuclear factor κ B and chemokine receptor
CXCR4 expression in patients with diffuse large B-cell lymphoma who
received rituximab-based therapy. Korean J. Intern. Med. 29, 785–792. doi:
10.3904/kjim.2014.29.6.785
Sierro, F., Biben, C., Martínez-Muñoz, L., Mellado, M., Ransohoff, R. M., Li, M.,
et al. (2007). Disrupted cardiac development but normal hematopoiesis in mice
deficient in the second CXCL12/SDF-1 receptor, CXCR7. Proc. Natl. Acad. Sci.
U.S.A. 104, 14759–14764. doi: 10.1073/pnas.0702229104
Sizemore, N., Leung, S., and Stark, G. R. (1999). Activation of Phosphatidylinositol
3-Kinase in Response to Interleukin-1 Leads to Phosphorylation and Activation
of the NF- κB p65/RelA Subunit. Mol. Cell. Biol. 19, 4798–4805.
Stiehl, T., and Marciniak-Czochra, A. (2012). Mathematical Modeling of
Leukemogenesis and Cancer Stem Cell Dynamics. Math. Model. Nat. Phenom.
7, 166–202. doi: 10.1051/mmnp/20127199
Sugiyama, T., Kohara, H., Noda, M., and Nagasawa, T. (2006). Maintenance
of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine
signaling in bone marrow stromal cell niches. Immunity 25, 977–988. doi:
10.1016/j.immuni.2006.10.016
Swaminathan, S., Klemm, L., Park, E., Papaemmanuil, E., Ford, A., Kweon, S.-M.,
et al. (2015). Mechanisms of clonal evolution in childhood acute lymphoblastic
leukemia. Nat. Immunol. 16, 766–774. doi: 10.1038/ni.3160
Tabe, Y., Jin, L., Tsutsumi-Ishii, Y., Xu, Y., McQueen, T., Priebe, W., et al. (2007).
Activation of integrin-linked kinase is a critical prosurvival pathway induced in
leukemic cells by bone marrow-derived stromal cells. Cancer Res. 67, 684–694.
doi: 10.1158/0008-5472.CAN-06-3166
Tabe, Y., and Konopleva, M. (2015). Role of microenvironment in resistance to
therapy in AML. Curr. Hematol. Malig. Rep. 10, 96–103. doi: 10.1007/s11899-
015-0253-6
Tapia-Abellán, A., Ruiz-Alcaraz, A. J., Antón, G., Miras-López, M., Francés, R.,
Such, J., et al. (2014). Regulatory role of PI3K-protein kinase B on the release of
interleukin-1β in peritoneal macrophages from the ascites of cirrhotic patients.
Clin. Exp. Immunol. 178, 525–536. doi: 10.1111/cei.12428
Tak, P. P., and Firestein, G. S. (2001). NF- κB: a key role in inflammatory diseases.
J. Clin. Invest. 107, 7–11. doi: 10.1172/JCI11830
Tarnowski, M., Liu, R., Wysoczynski, M., Ratajczak, J., Kucia, M., and Ratajczak,
M. Z. (2010). CXCR7: a new SDF-1-binding receptor in contrast to normal
CD34+ progenitors is functional and is expressed at higher level in
human malignant hematopoietic cells. Eur. J. Haematol. 85, 472–483. doi:
10.1111/j.1600-0609.2010.01531.x
Tesio, M., Oser, G. M., Baccelli, I., Blanco-Bose, W., Wu, H., Göthert, J. R.,
et al. (2013). Pten loss in the bone marrow leads to G-CSF-mediated HSC
mobilization. J. Exp. Med. 210, 2337–2349. doi: 10.1084/jem.20122768
Tokoyoda, K., Egawa, T., Sugiyama, T., Choi, B.-I., and Nagasawa, T. (2004).
Cellular niches controlling B lymphocyte behavior within bone marrow during
development. Immunity 20, 707–718. doi: 10.1016/j.immuni.2004.05.001
Tomasetti, C., and Vogelstein, B. (2015). Variation in cancer risk among tissues
can be explained by the number of stem cell divisions. Science 347, 78–81. doi:
10.1126/science.1260825
Torossian, F., Anginot, A., Chabanon, A., Clay, D., Guerton, B., Desterke, C., et al.
(2014). CXCR7 participates in CXCL12-induced CD34+ cell cycling through
b-arrestin–dependent Akt activation. Blood 123, 191–202. doi: 10.1182/blood-
2013-05-500496
Tzeng, Y.-S., Li, H., Kang, Y.-L., Chen, W.-C., Cheng, W.-C., and Lai, D.-M.
(2011). Loss of Cxcl12/Sdf-1 in adult mice decreases the quiescent state of
hematopoietic stem/progenitor cells and alters the pattern of hematopoietic
regeneration after myelosuppression. Blood 117, 429–439. doi: 10.1182/blood-
2010-01-266833
Frontiers in Physiology | www.frontiersin.org
14
August 2016 | Volume 7 | Article 349
Enciso et al.
Modeling CXCR4/CXCL12 Disruption in Acute Leukemia
Ueda, Y., Yang, K., Foster, S. J., Kondo, M., and Kelsoe, G. (2004). Inflammation
controls B lymphopoiesis by regulating chemokine CXCL12 expression. J. Exp.
Med. 199, 47–58. doi: 10.1084/jem.20031104
Uto-Konomi, A., McKibben, B., Wirtz, J., Sato, Y., Takano, A., Nanki, T.,
et al. (2013). CXCR7 agonists inhibit the function of CXCL12 by down-
regulation of CXCR4. Biochem. Biophys. Res. Commun. 431, 772–776. doi:
10.1016/j.bbrc.2013.01.032
Vadillo, E., Dorantes-Acosta, E., Arriaga-Pizano, L., Chavez-Gonzalez, A., Reyes-
Maldonado, E., Garrett, K. P., et al. (2014). Adult, but not neonatal, human
lymphoid progenitors respond to TLR9 ligation by producing functional
NK-like cells. Exp. Hematol. 42, 562–573. doi: 10.1016/j.exphem.2014.
03.008
Vadillo,
E.,
Dorantes-acosta,
E.,
and
Pelayo,
R.
(2013).
“Regulation
of
hematopoietic stem / progenitor cell development by inflammation cues,” in
Molecular Aspects of Inflammation, eds L. Perez-Martinez, G. Pedraza-Alva,
and E. F. Osorio (Kerala: Research Signpost), 71–87.
Vagima, Y., Avigdor, A., Goichberg, P., Shivtiel, S., Tesio, M., Kalinkovich, A.,
et al. (2009). MT1-MMP and RECK are involved in human CD34+ progenitor
cell retention, egress, and mobilization. J. Clin. Invest. 119, 492–503. doi:
10.1172/JCI36541
van den Berk, L. C. J., van der Veer, A., Willemse, M. E., Theeuwes, M. J. G., a,
Luijendijk, M. W., Tong, W. H., et al. (2014). Disturbed CXCR4/CXCL12 axis
in paediatric precursor B-cell acute lymphoblastic leukaemia. Br. J. Haematol.
166, 240–249. doi: 10.1111/bjh.12883
Vilchis-Ordoñez, A., Contreras-Quiroz, A., Vadillo, E., Dorantes-Acosta, E., Reyes-
López, A., Quintela-Nuñez del Prado, H. M., et al. (2015). Bone marrow cells
in acute lymphoblastic leukemia create a proinflammatory microenvironment
influencing normal hematopoietic differentiation fates. Biomed Res. Int.
2015:386165. doi: 10.1155/2015/386165
Vilchis-Ordoñez, A., Dorantes-Acosta, E., Vadillo, E., López-Martínez, B., and
Pelayo, R. (2016). “Early hematopoietic differentiation in acute lymphoblastic
leukemia: the interplay between leukemia-initiating cells and abnormal bone
marrow microenvironment,” in Etiology of Acute Leukemias in Children ed J.
M. Mejía-Aranguré (Cham: Springer International publishing), 291–318.
Wang, L., You, L., Ni, W., Ma, Q., Tong, Y., Mao, L., et al. (2013). β-Catenin and
AKT are promising targets for combination therapy in acute myeloid leukemia.
Leuk. Res. 37, 1329–1340. doi: 10.1016/j.leukres.2013.06.023
Wang, M. W., Consoli, U., Lane, C. M., Durett, A., Lauppe, M. J., Champlin,
R., et al. (1998). Rescue from apoptosis in early (CD34-selected) versus
late (non-CD34-selected) human hematopoietic cells by very late antigen 4-
and vascular cell adhesion molecule (VCAM) 1-dependent adhesion to bone
marrow stromal cells. Cell Growth Differ. 9, 105–112.
Wang, T., Zou, J., Cunningham, C., and Secombes, C. J. (2002). Cloning and
functional characterisation of the interleukin-1 β1 promoter of rainbow
trout (Oncorhynchus mykiss). Biochim. Biophys. Acta 1575, 108–116. doi:
10.1016/S0167-4781(02)00235-X
Wang, X., Cheng, Q., Li, L., Wang, J., Xia, L., Xu, X., et al. (2012). Toll-like receptors
2 and 4 mediate the capacity of mesenchymal stromal cells to support the
proliferation and differentiation of CD34+ cells. Exp. Cell Res. 318, 196–206.
doi: 10.1016/j.yexcr.2011.11.001
Wang, Z. J., Zhang, F. M., Wang, L. S., Yao, Y. W., Zhao, Q., and Gao, X.
(2009). Lipopolysaccharides can protect mesenchymal stem cells (MSCs) from
oxidative stress-induced apoptosis and enhance proliferation of MSCs via
Toll-like receptor(TLR)-4 and PI3K/Akt. Cell Biol. Int. 33, 665–674. doi:
10.1016/j.cellbi.2009.03.006
Welner, R. S., Pelayo, R., Nagai, Y., Garrett, K. P., Wuest, T. R., Carr, D. J.,
et al. (2008). Lymphoid precursors are directed to produce dendritic cells as
a result of TLR9 ligation during herpes infection. Blood 112, 3753–3761. doi:
10.1182/blood-2008-04-151506
Williams, D. L., Li, C., Ha, T., Ozment-Skelton, T., Kalbfleisch, J. H., Preiszner,
J., et al. (2004). Modulation of the phosphoinositide 3-kinase pathway alters
innate resistance to polymicrobial sepsis. J. Immunol. 172, 449–456. doi:
10.4049/jimmunol.172.1.449
Wu, M., Yang, X., and Chan, C. (2009). A dynamic analysis of IRS-PKR
signaling in liver cells: a discrete modeling approach. PLoS ONE 4:e8040. doi:
10.1371/journal.pone.0008040
Xu, J., Qian, J., Xie, X., Lin, L., Zou, Y., Fu, M., et al. (2012). High density
lipoprotein protects mesenchymal stem cells from oxidative stress-induced
apoptosis via activation of the PI3K/Akt pathway and suppression of reactive
oxygen species. Int. J. Mol. Sci. 13, 17104–17120. doi: 10.3390/ijms131217104
Yamazaki, S., Iwama, A., Takayanagi, S., Morita, Y., Eto, K., Ema, H., et al.
(2006). Cytokine signals modulated via lipid rafts mimic niche signals and
induce hibernation in hematopoietic stem cells. EMBO J. 25, 3515–3523. doi:
10.1038/sj.emboj.7601236
Yang, D., Elner, S. G., Bian, Z. M., Till, G. O., Petty, H. R., and Elner, V. M.
(2007). Pro-inflammatory cytokines increase reactive oxygen species through
mitochondria and NADPH oxidase in cultured RPE cells. Exp. Eye Res. 85,
462–472. doi: 10.1016/j.exer.2007.06.013
Yi, L., Chandrasekaran, P., and Venkatesan, S. (2012). TLR signaling paralyzes
monocyte chemotaxis through synergized effects of p38 MAPK and global
rap-1 activation. PLoS ONE 7:30404. doi: 10.1371/journal.pone.0030404
Zeng, H., Yücel, R., Kosan, C., Klein-Hitpass, L., and Möröy, T. (2004).
Transcription
factor
Gfi1
regulates
self-renewal
and
engraftment
of
hematopoietic stem cells. EMBO J. 23, 4116–4125. doi: 10.1038/sj.emboj.
7600419
Zhang, X., Liu, Y., Si, Y., Chen, X., Li, Z., Gao, L., et al. (2012). Effect of Cx43 gene-
modified leukemic bone marrow stromal cells on the regulation of Jurkat cell
line in vitro. Leuk. Res. 36, 198–204. doi: 10.1016/j.leukres.2011.10.001
Zhang, Y., Zhai, W., Zhao, M., Li, D., Chai, X., Cao, X., et al. (2015). Effects
of Iron overload on the bone marrow microenvironment in mice. PLoS ONE
10:e0120219. doi: 10.1371/journal.pone.0120219
Zhuang, D., Qiu, Y., Kogan, S. C., and Dong, F. (2006). Increased CCAAT
enhancer-binding protein ε (C/EBPε) expression and premature apoptosis in
myeloid cells expressing Gfi-1 N382S mutant associated with severe congenital
neutropenia. J. Biol. Chem. 281, 10745–10751. doi: 10.1074/jbc.M510924200
Conflict of Interest Statement: The authors declare that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2016 Enciso, Mayani, Mendoza and Pelayo. This is an open-access
article distributed under the terms of the Creative Commons Attribution License (CC
BY). The use, distribution or reproduction in other forums is permitted, provided the
original author(s) or licensor are credited and that the original publication in this
journal is cited, in accordance with accepted academic practice. No use, distribution
or reproduction is permitted which does not comply with these terms.
Frontiers in Physiology | www.frontiersin.org
15
August 2016 | Volume 7 | Article 349
|
27594840
|
PI3KAkt_H = ( ( TLR_H ) AND NOT ( FoxO3a_H ) ) OR ( ( CXCR4_H AND ( ( ( CXCR7_H ) ) ) ) AND NOT ( FoxO3a_H ) ) OR ( ( VLA4_H ) AND NOT ( FoxO3a_H ) ) OR ( ( GCSF ) AND NOT ( FoxO3a_H ) ) OR ( ( ROS_H ) AND NOT ( FoxO3a_H ) )
CXCR4_H = ( ( ( ( CXCL12_M ) AND NOT ( CXCR7_H ) ) AND NOT ( GCSF ) ) AND NOT ( Gfi1_H ) )
TLR_M = ( lTLR )
GSK3B_M = NOT ( ( PI3KAkt_M ) )
ROS_H = ( ( IL1 AND ( ( ( TLR_H ) ) ) ) AND NOT ( FoxO3a_H ) )
FoxO3a_H = ( ( ( Bcatenin_H ) AND NOT ( PI3KAkt_H ) ) AND NOT ( ERK_H ) ) OR ( ( ( ROS_H ) AND NOT ( PI3KAkt_H ) ) AND NOT ( ERK_H ) )
ROS_M = ( ( IL1 AND ( ( ( TLR_M ) ) ) ) AND NOT ( FoxO3a_M ) )
ERK_H = ( ( ( CXCR4_H AND ( ( ( PI3KAkt_H ) ) ) ) AND NOT ( FoxO3a_H ) ) AND NOT ( GSK3B_H ) ) OR ( ( ( VLA4_H ) AND NOT ( FoxO3a_H ) ) AND NOT ( GSK3B_H ) ) OR ( ( ( Gfi1_H ) AND NOT ( FoxO3a_H ) ) AND NOT ( GSK3B_H ) ) OR ( ( ( CXCR7_H ) AND NOT ( FoxO3a_H ) ) AND NOT ( GSK3B_H ) ) OR ( ( ( GCSF ) AND NOT ( FoxO3a_H ) ) AND NOT ( GSK3B_H ) ) OR ( ( ( ROS_H ) AND NOT ( FoxO3a_H ) ) AND NOT ( GSK3B_H ) )
lTLR = ( lTLR )
VCAM1_M = ( ( NfkB_M ) OR ( PI3KAkt_M ) ) OR NOT ( PI3KAkt_M OR Bcatenin_M OR NfkB_M )
NfkB_H = ( ( TLR_H ) AND NOT ( FoxO3a_H ) ) OR ( ( ROS_H ) AND NOT ( FoxO3a_H ) ) OR ( ( IL1 AND ( ( ( PI3KAkt_H ) ) ) ) AND NOT ( FoxO3a_H ) )
FoxO3a_M = ( ( ( ROS_M ) AND NOT ( PI3KAkt_M ) ) AND NOT ( ERK_M ) ) OR ( ( ( Bcatenin_M ) AND NOT ( PI3KAkt_M ) ) AND NOT ( ERK_M ) )
Cx43_M = ( Cx43_M )
VLA4_H = ( VCAM1_M AND ( ( ( CXCR4_H ) ) ) )
NfkB_M = ( ROS_M AND ( ( ( ERK_M ) ) ) ) OR ( TLR_M ) OR ( IL1 AND ( ( ( PI3KAkt_M ) ) ) )
CXCR7_H = ( CXCL12_M AND ( ( ( NfkB_H ) ) ) )
Bcatenin_H = NOT ( ( GSK3B_H ) )
GSK3B_H = NOT ( ( PI3KAkt_H ) )
Gfi1_H = ( ( GCSF ) AND NOT ( Gfi1_H ) ) OR ( ( TLR_H ) AND NOT ( Gfi1_H ) )
ERK_M = ( ROS_M ) OR ( GCSF ) OR ( TLR_M )
GCSF = ( IL1 )
IL1 = ( ( NfkB_M ) AND NOT ( PI3KAkt_M ) ) OR ( ( ROS_M ) AND NOT ( PI3KAkt_M ) ) OR ( ( NfkB_H ) AND NOT ( PI3KAkt_H ) ) OR ( ( ROS_H ) AND NOT ( PI3KAkt_H ) )
TLR_H = ( lTLR )
CXCL12_M = ( ( ( ( Cx43_M ) AND NOT ( Bcatenin_M ) ) AND NOT ( NfkB_M ) ) AND NOT ( GCSF ) )
PI3KAkt_M = ( ROS_M ) OR ( GCSF ) OR ( TLR_M )
Bcatenin_M = NOT ( ( NfkB_M ) OR ( FoxO3a_M ) OR ( GSK3B_M ) )
|
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
DOI 10.1186/s12859-017-1522-2
RESEARCH
Open Access
Towards targeted combinatorial therapy
design for the treatment of castration-resistant
prostate cancer
Osama Ali Arshad1,2 and Aniruddha Datta1,2*
From Third International Workshop on Computational Network Biology: Modeling, Analysis, and Control (CNB-MAC 2016)
Seattle, WA, USA. 02-Oct-16
Abstract
Background: Prostate cancer is one of the most prevalent cancers in males in the United States and amongst the
leading causes of cancer related deaths. A particularly virulent form of this disease is castration-resistant prostate
cancer (CRPC), where patients no longer respond to medical or surgical castration. CRPC is a complex, multifaceted
and heterogeneous malady with limited standard treatment options.
Results: The growth and progression of prostate cancer is a complicated process that involves multiple pathways.
The signaling network comprising the integral constituents of the signature pathways involved in the development
and progression of prostate cancer is modeled as a combinatorial circuit. The failures in the gene regulatory network
that lead to cancer are abstracted as faults in the equivalent circuit and the Boolean circuit model is then used to
design therapies tailored to counteract the effect of each molecular abnormality and to propose potentially
efficacious combinatorial therapy regimens. Furthermore, stochastic computational modeling is utilized to identify
potentially vulnerable components in the network that may serve as viable candidates for drug development.
Conclusion: The results presented herein can aid in the design of scientifically well-grounded targeted therapies that
can be employed for the treatment of prostate cancer patients.
Keywords: Prostate cancer, Gene regulatory networks, Boolean modeling, Combination therapy, Stochastic logic,
Vulnerability assessment
Background
Prostate cancer is the most common noncutaneous male
malignancy and one of the leading causes of cancer mor-
tality in the western world [1]. The growth and pro-
gression of prostate cancer is stimulated by androgens
[2]. Androgens are male sex steroid hormones that are
responsible for the development of male characteristics.
Testosterone is the most important androgen in men. The
effects of androgens are mediated through the androgen
receptor (AR) [3]. The androgen receptor is a nuclear
*Correspondence: datta@ece.tamu.edu
1Department of Electrical and Computer Engineering, Texas A&M University,
College Station, TX, USA
2Center for Bioinformatics and Genomics Systems Engineering, Texas A&M
University, College Station, TX, USA
receptor, which is activated in response to the binding
of androgens. Upon activation, it mediates transcription
of target genes that modulate growth and differentia-
tion of prostate epithelial cells. In malignant prostate
cells, androgen signaling is deregulated and the homeo-
static balance between the rate of cell proliferation and
programmed cell death is lost. As prostate cancer relies
on androgens for growth, the main line of treatment
focuses on abrogating the action of androgens. Andro-
gen deprivation therapy (ADT) in the form of surgical
or medical castration is the cornerstone of treatment for
prostate cancer [4]. Initially, androgen ablation induces
significant regression of the tumor. However, the response
to ADT is temporary and prostate cancer invariably
stops responding to this treatment regimen, leading to a
© The Author(s). 2017 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 6 of 59
clinical condition that is known as hormone-refractory
prostate cancer, androgen-independent prostate cancer or
castration-resistant prostate cancer (CRPC). CRPC is a
more aggressive and typically lethal phenotype where the
tumor continues to grow in spite of the very low levels
(<50 ng/ml) of circulating serum testosterone. Standard
treatment options are limited and palliative docetaxel-
based chemotherapy is generally used for patients who
have become refractory to hormone treatment. How-
ever, median survival time for patients following first-line
chemotherapeutic treatment is just eighteen to twenty-
four months [5]. There is therefore a clear rationale for
advances in alternative therapeutics in order to evolve and
expand the landscape of treatment options for malignant
forms of prostate cancer that recur after abatement.
Over recent years, there has been a significant effort
towards furthering our understanding of the molecular
mechanisms underpinning tumor development, growth
and progression. It is now appreciated that in spite of
castrate levels of androgens, the cancer cells are able to
maintain persistent androgen receptor signaling through
a variety of contributory mechanisms including AR gene
amplification that results in overexpression of AR, gain-
of-function mutations in AR which enable promiscuous
activation of the receptor through other steroids or even
in the absence of ligand binding, changes in AR co-
activators and the expression of AR splice variants [6].
This compensatory response allows cancer cells to sur-
vive in a low testosterone environment and the reactivated
AR signaling axis continues to play a role after neo-
plastic transformation. Additionally, certain androgen-
independent cellular signaling pathways that promote
proliferation and inhibit apoptosis, have been critically
implicated as drivers of continued progression of prostate
cancer. Hence, accumulating evidence indicates that the
growth and progression of prostate cancer is a compli-
cated process that involves interaction between multiple
pathways. Advances in our knowledge of the biology of
prostate cancer has led to the development of a number
of novel therapies designed to target signaling pathways
involved in disease progression. With the exception of cer-
tain androgen synthesis and AR signaling antagonists that
have received regulatory approval, these advanced agents
are under various stages of clinical trials [7].
Castration-resistant prostate cancer is a complex mal-
ady. Given the inherent complexity of the CRPC signal-
ing cascade, there is no one dominant molecular driver
across all tumors and hence no single drug can act as
a “magic bullet” by being uniformly effective for treat-
ing the malignancy [8, 9]. At best, limited benefit will be
derived from targeting a single molecule. Rational com-
binations of signal-modulating therapeutic agents have
higher likelihood of yielding better outcomes. While there
are several drugs being tested on cell lines, most of these
studies focus on a single pharmaceutical agent and very
few of those experiments involve trying out drug com-
binations. Furthermore, prostate cancer is a markedly
heterogeneous disease, with different tumors varying in
their composition and makeup. In other words, different
tumors will harbor different malfunctions in the signaling
pathways. Thus, tailored targeted therapies based on indi-
vidual tumor characteristics are required to maximize the
potential benefits from treatment.
Mathematical and computational modeling plays a piv-
otal role in systems biology in elucidating biological
insights from large-scale biomolecular signaling networks
that are not amenable to straightforward intuitive inter-
pretation. A diverse array of formalisms have been pro-
posed in this domain as suitable representations for
complex multicomponent networks such as cellular
signaling pathways [10]. Amongst these frameworks,
Boolean network models [11, 12] have emerged as
an extremely useful parameter-free approach to cap-
ture the qualitative behavior of extensive genetic net-
works wherein knowledge of kinetic parameters is scarce.
Boolean logic models have been successfully applied to
study biological signaling networks and cellular processes
[13, 14], for instance the cell cycle [15], apoptosis [16],
the T cell survival network [17], hypoxia stress response
pathways [18] and the gene regulatory network regulat-
ing cortical area development [19]. In this paper, we use
Boolean logic modeling of the key signaling pathways
implicated in the development and progression of prostate
cancer to simultaneously test various combinations of
agents for their efficacy in attenuating cancer growth and
design targeted therapies for the management of the dis-
ease. In addition, we attempt to delineate components
in the signaling network that can be pharmacologically
manipulated to therapeutic advantage.
Methods
Prostate cancer signal transduction network
Cellular processes such as growth and division are regu-
lated by an interconnected network of molecules referred
to as signaling pathways. Key cellular signal transduc-
tion pathways known to play a major role in cell survival,
growth, differentiation and the development of castration-
resistance in prostate cancer are the Androgen Recep-
tor (AR), PI3K/AKT/mTOR and Mitogen-Activated
Protein Kinase (MAPK) pathways. The aberrant behavior
of prostate cancer cells is characterized by dysfunction in
these selective oncogenic signaling pathways promoting
malignant characteristics. These pathways play a role in a
diverse range of essential physiological cellular processes
such as differentiation, survival, proliferation, protein
synthesis and metabolism. Malfunctions in these path-
ways are common in prostate cancer malignancies. For
example, approximately 70% of advanced prostate cancers
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 7 of 59
have genomic alterations in the PI3K/AKT/mTOR path-
way [20]. These three pathways are the most frequently
over-activated pathways increasing survival of cancer cells
and promoting cancer progression [21]. A schematic
representation of these pathways is shown in Fig. 1
[22–24]. The pharmacologic agents depicted in red boxes
in the figure are highly specific pathway inhibitors. These
reagents modulate growth-factor receptors and the down-
stream pathways abnormally activated in CRPC by target-
ing with great specificity certain signaling nodes in the
network.
Boolean modeling of prostate cancer signaling
In the context of methodologies that are applied to model
cellular signal transduction networks, Boolean networks
are probably the simplest where the state of each node
in the network is either active (on) or inactive (off). In a
Boolean network, the nodes are the genes and the edges
represent the interaction amongst the genes. Since the
molecules in a gene-regulatory-network (GRN) exhibit
switch-like behavior, genes may be regarded as binary
devices where a gene can be considered to be active if it
is being transcribed and inactive if it is not. Moreover,
the relationships amongst the genes may be represented
by means of logical functions. Thus, a GRN is amenable
to such a representation. The Boolean formalism is anal-
ogous to a digital circuit where logic gates can be used to
represent the regulatory relationships amongst the nodes
and the activation level of the nodes is indicated by binary
logic. The biological interactions amongst the various
nodes (genes) represented in the gene regulatory net-
work of Fig. 1 can therefore be translated to an equivalent
Boolean circuit [25]. Let’s say either gene X or Y can acti-
vate a third gene Z, then we can model this component
of the signaling network with an OR gate with two inputs,
namely X and Y and with output Z. Thus, the signaling
network of Fig. 1 can be mapped to the combinational cir-
cuit shown in Fig. 2. This digital logic circuit represents
our multi-input multi-output (MIMO) systems model of
the prostate cancer signaling transduction network.
Cancer is a disease of abnormal cell signaling caused by
a breakdown in the normal signaling pathways leading to
the loss of cell cycle control and uncontrolled cell prolifer-
ation. These abnormalities in the signaling network can be
represented as stuck-at faults [26]. A stuck-at fault is said
to occur when a line in the network is permanently set to
a fixed value of one (stuck-at-one fault) or zero (stuck-at-
zero fault) with the result that the state of the line is stuck
at the faulty value and no longer depends on the state
of the signaling network upstream that drives that line
Fig. 1 Prostate cancer signal transduction network. A schematic diagram of key signaling pathways deregulated in prostate cancer. Black and red
lines represent activating and inhibiting interactions respectively whereas the red boxes depict prostate cancer drugs at their corresponding points
of intervention in the network
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 8 of 59
Androgens
(13)
NKX3.1
(7)
PTEN
IGF
NRG1
HBEGF
EGF
(3)
IGFR1A/B
(2)
EFGR
(1) EGFR
ERBB2
(4)
ERBB2/3
(6) Ras
(5)
GRB2/
SOS
(8) IRS1
(9)
PIK3CA
(10) PIP3
(11)
PDPK1
(16) Raf
(17)
MEK1
(19)
TSC1/2
(20)
RHEB
(12) AKT
(21)
mTOR
(18)
ERK1/2
SP1
SRF-ELK1
(15)
AR-AR
TMPRSS2
PSA
BCL2
CDK2-
Cyclin E
(24)
p21
(23)
BAD
(22)
RP6SKB1
Enzalutamide
AZD5363
Temsirolimus
BKM120
Cixutumumab
Lapatinib
AZD6244
(14) AR/HSP
Fig. 2 Boolean model. Combinational circuit model of prostate cancer signaling pathways. Each node is assigned a numeric label in parentheses.
These labels also serve to enumerate the fault locations with stuck-at-one and stuck-at-zero faults in black and red numerals respectively. The
dotted arrows indicate the intervention points for the respective drugs
i.e. the faulty line has a constant (1/0) value independent
of other signal values in the circuit. A stuck-at-fault can
occur either at the input or output of a gate. An example
of a stuck-at-fault is given in Fig. 3. Suppose the input vec-
tor is <abcd>= 1100. In this case, the output is 0. However,
if there is a stuck-at-one fault at the output of the NAND
gate with the same input vector as before, the output of the
faulty circuit is one instead of zero. This notion of stuck-
at-faults has immediate biological relevance: on account
of mutations or other structural abnormalities, a gene
might become dysfunctional and hence stuck at a partic-
ular state irrespective of the signals that it is receiving
from surrounding genes [27]. These biological defects can
be abstracted as stuck-at faults. For instance, as discussed
earlier, a diverse array of mechanisms engender persistent
AR signaling in CRPC even with castrate serum levels of
androgen. This constitutive (permanent) activation of the
androgen receptor where the receptor remains active i.e.
continues to signal downstream even in the absence of
androgens can be represented as a stuck-at-1 fault. By the
same token, the inactivation in cancer of a tumor suppres-
sor, which acts as a molecular brake on cell growth in a
normal cell, can be represented as a stuck-at-0 fault. From
our Boolean circuit model, we can explicitly enumerate
the different locations where a fault can occur. These fault
locations are numbered in Fig. 2 with the stuck-at-0 and
1
X
a = 1
b = 1
c = 0
d = 0
0/1
Fig. 3 Circuit with stuck-at fault. An example of a stuck-at fault. In the absence of the stuck-at fault, the output is zero. If there is a stuck-at-one fault
at the location marked with a cross, the output of the faulty circuit becomes one
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 9 of 59
stuck-at-1 faults in red and black numerals respectively.
There is a total number of 24 possible fault locations.
The objective is to counteract the effect of these faults
by targeted drug intervention, so we incorporate the drugs
in our model. The drug intervention points are illustrated
in Fig. 2 which are the locations of the molecules that
these prostate cancer drugs are known to target. Since
the drugs inhibit the activity of their target i.e. the main
mechanism of action of the anti-cancer drugs is to cut off
downstream signaling, their action is incorporated in our
model as an inverted input to an AND gate with the result
that whenever the drug is applied, the gene that it targets is
turned off.
Simulation for fault mitigation with drug intervention
We can now use our Boolean model to test different com-
bination therapies in terms of their efficacy in mitigating
the effects of the faults. For each fault, we would like to
intervene with the best possible drug combination i.e. we
want to determine which set of drugs would be most effec-
tive in attempting to nullify the effect of that fault, thereby
providing us with a targeted therapy based on the tumor
signature. Define, the input vector as follows:
INPUT =
EGF, HBEGF, IGF, NRG1, PTEN, NKX3. 1, Androgens
The first four components of this vector are growth fac-
tors, which are external signals that stimulate a cell to
grow and replicate. The next two input components,
namely PTEN and NKX3.1 are tumor suppressors which
act as molecular brakes on cell division. The last input
vector component consists of the external hormones that
stimulate the AR pathway in a normal prostate cell. The
input vector is set to be [0000110]. This corresponds to all
the external signals that stimulate cell growth being absent
and the molecular brakes being active i.e. this input vector
corresponds to a non-proliferative input which produces a
non-proliferative output in the fault-free case. The output
vector is defined to be:
OUTPUT =
SP1, SRF-ELK1, PSA, TMPRSS2, BCL2, CDK2-CyclinE
The output vector consists of key markers of cell growth
and proliferation in prostate cancer. In the fault-free sce-
nario, a non-proliferative input to the regulatory network
should produce a non-proliferative output characterized
by the all-zero vector. However, faults in the network will
produce a non-zero (proliferative) output even when the
input is non-proliferative. The objective is to drive the
faulty network’s output as close as possible to that of the
fault-free circuit i.e. towards the all-zero vector through
targeted drug intervention. Define, the drug vector as:
DRUG VECTOR =
Lapatinib, Cixutumumab, AZD6244, BKM120, AZD5363,
Temsirolimus, Enzalutamide
Each component of the drug vector is one if the corre-
sponding drug is applied and is zero otherwise i.e. the
ith bit of the drug vector is one if the drug is selected
and zero if it is not. Thus, for example, the drug vec-
tor [0010010] represents the combination of AZD6244
and Temsirolimus. Since, the total number of drugs is
seven, the number of possible drug combinations is 128.
The objective is to determine the best possible therapy
for each fault. Each fault represents a different molecular
abnormality and hence a tumor with a different profile.
For each of the faults, the problem is to find the drug
selection that can rectify the fault i.e. change the faulty
output to the correct output. If that is not possible, the
best drug vector will drive the output as close as pos-
sible to the fault-free output. A simple metric that can
be used as a distance measure to determine how far the
output vector is from the fault-free vector is Hamming
distance. Faults that produce an output vector with a
greater Hamming distance from the correct output have
more of the proliferative genes active and presumably a
greater proliferative effect. Since the correct output is the
all-zero vector, the Hamming distance of the output vector
from the correct output is simply the Hamming weight of
the output vector (for binary vectors Hamming weight is
equivalent to the L1-norm). For each fault, we determine
the output under every possible drug vector. The best
therapy for that fault is the drug vector that produces the
output with the smallest Hamming weight. In addition,
since the drugs have deleterious side-effects, we would
like to choose a drug combination with the fewest number
of drugs. Thus, the best targeted therapy for each of the
cancer-inducing faults is the one that under the presence
of the fault, produces the best output with the smallest
Hamming weight with the minimal number of drugs.
To determine the best combination therapy across all
faults, for each drug combination we determine the sum
of the Hamming weights of the output vector across all
possible combinations of faults and choose the drug com-
bination that yields the smallest total. In order to keep the
computation tractable, we restrict the number of possible
faults in any fault combination to be no more than three
i.e. up to three genes can be faulty simultaneously. We
constrain the cardinality of the drug vector to be less than
or equal to three, in essence limiting the number of drugs
in the combination to three since on account of the harm-
ful side-effects of the drugs, administering four or more
cancer drugs simultaneously might not be prudent.
Let us formalize the qualitative description above of the
selection of best therapy for each fault and that of the
overall optimal drug vector. For the Boolean network (BN)
of Fig. 2, let N, M and P be the total number of primary
inputs, primary outputs and fault locations respectively,
then N=7, M=6 and P=24. Let x ∈X and z ∈Z be
the input and output vectors respectively where X and Z
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 10 of 59
represent the space of all binary vectors of dimensions N
and M respectively. Let x∗=[0, 0, 0, 0, 1, 1, 0] be the input
vector corresponding to the non-proliferative input.
Let D represent the total number of drug combinations
(vectors) with no more than three drugs in any combina-
tion, then D =
3
k=0
7
k
. Denote each drug vector in the
drug space as diwith i = 0, . . . , D −1 (d0 is the all-zero
drug vector meaning no drug is applied). Let D be this
space of drug vectors.
Let C be the total number of fault combinations with
no more than three faults in any combination, then C =
3
k=0
P
k
. Assign each fault combination in the fault space a
label fj with j = 0, . . . , C −1 (f0 represents the fault-free
case). Let F be this set of faults.
Let ψ denote the mapping from a given input vector,
drug combination and fault combination to an output vec-
tor: x ∈X , d ∈D, f ∈F
ψ−→z ∈Z i.e. ψ represents the
output of the BN for a given input x when a drug combi-
nation d is applied under fault scenario f . Let ψi be the ith
component of this M-dimensional vector ψ.
The best drug vector di, i ∈{0, 1, . . . , D −1} for each
single fault fj, j ∈{1, 2, . . . , P} is the vector of smallest
Hamming weight that minimizes
ψ
x∗, di, fj
1.
The optimal drug combination across all faults is:
d∗
i = arg min
di
C−1
j=1
ψ
x∗, di, fj
1
(1)
d∗
i is determined by exhaustive enumeration by explic-
itly searching for the drug combination that for a non-
proliferative input, minimizes the sum of Hamming
weights (L1-norms) of the output vector across all possible
combinations of faults.
Node vulnerability assessment
In electronic circuits, reliability refers to the probability of
a circuit functioning as intended i.e. producing the cor-
rect output. Reliability assessment is used to determine
the vulnerability of a circuit to faults. A number of differ-
ent techniques have been proposed for reliability analysis
in digital circuits [28]. Recently, in [29] a scalable, effi-
cient and accurate simulation-based framework based on
stochastic computations was introduced for logic circuit
reliability evaluation. In biological systems, dysfunctions
in nodes in the signaling network cause deviation from
normative behavior. Reliability assessment methodologies
can be leveraged on Boolean network models of pathways
to determine the vulnerability of the network to the dys-
function of each node [30, 31]. In this section we conduct a
stochastic logic based vulnerability analysis of the prostate
cancer signal transduction network in order to discover
the most vulnerable nodes thereby allowing us to priori-
tize such segments in the network whose perturbation has
the greatest potential to yield the most clinical benefit.
In stochastic logic, signal probabilities are encoded in
random binary bit streams (the signal probability of a node
corresponds to the likelihood of that node having logic
value one). For example, the binary sequence 0110010100
of length ten encodes the probability 0.4 since the propor-
tion of ones in this sequence is 4
10. In practice, the length
of the stochastic sequences typically used is much larger.
Since the biological literature is devoid of precise lig-
and binding probabilities, each primary input is assumed
equally likely to be 0 or 1 i.e. all primary input signal
probabilities are taken to be 0.5.
Stochastic logic often makes use of Bernoulli sequences
for the random binary streams where each bit in the
stream is generated independently from a Bernoulli ran-
dom variable with a probability of one equal to p. The use
of probabilistic sequences inevitably introduces stochas-
tic fluctuations which implies that the result produced
is non-deterministic. These fluctuations can be signifi-
cantly reduced by representing the initial input proba-
bilities by non-Bernoulli sequences [32] defined as ran-
dom permutations of sequences containing a fixed num-
ber of ones and zeros. For a given probability p and
sequence length L, a non-Bernoulli sequence contains a
fixed number pL of ones, with the positions of the ones
determined by a random permutation. Thus, for exam-
ple, to represent the probability 0.5 by a non-Bernoulli
stream of length 10, we could randomly permute the
sequence 1111100000 which has five ones (instead of
generating each bit from a Bernoulli random variable
with p = 0.5 as would have been done to represent
the same probability by a Bernoulli sequence). We use
non-Bernoulli sequences of random permutations of fixed
number of ones and zeros in order to encode the initial
input probabilities.
A logic circuit operating on stochastic bit streams
(see Fig. 4 for an example), accepts as input random
sequences representing the probability of each input being
one and produces ones and zeros like any digital circuit
[33] i.e. a stochastic logic circuit uses Boolean gates to
operate on sequences of random bits. Each bit-stream
represents a stochastic number interpreted as the proba-
bility of seeing a one in an arbitrary position. Thus, the
computations performed by such a circuit are probabilis-
tic in nature. The output bit stream produced can be
decoded as the probability of the output being one by
counting the number of ones in the stream and dividing by
its length.
The vulnerability of a node is defined as the proba-
bility that the system produces incorrect output if that
particular node is dysfunctional (faulty) i.e. it is the proba-
bility that the output of the network is different when that
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 11 of 59
0001111010
p1 = 0.5
1110000101
0111111101
p2 = 0.8
0110000101
0010100111
p3 = 0.5
0110100111
pout = 0.6
Fig. 4 A stochastic logic circuit. An example of a stochastic logic circuit
node is dysfunctional and is the complement of reliability.
The procedure to determine the node vulnerabilities is
illustrated in Fig. 5 is as follows. We generate non-
Bernoulli sequences of length L=1,000,000 in which
exactly half of the bits are set to one at each of the seven
initial inputs. The input stochastic sequences are propa-
gated through both the original error-free circuit and the
circuit in which the node of interest is dysfunctional. As
discussed in the previous section, the dysfunction of a
node is represented by a corresponding stuck-at fault of
the requisite type at the particular location. This produces
two sets of stochastic bit streams, one at each of the pri-
mary outputs of the fault-free circuit and the other at the
primary outputs of the unreliable circuit. The proportion
of ones in the output bit stream encodes the output signal
probabilities i.e. the probability of the output being one.
Since the reliability of the circuit under the fault is the
probability that the circuit output is same as that of the
fault-free circuit, the sequence encoding the output reli-
ability can be obtained from the output sequence of the
faulty circuit by comparing it to the output sequence of the
fault-free circuit and setting each bit to one whenever the
corresponding bits in the sequences are the same and zero
if they are different. The proportion of ones in this result-
ing sequence will then correspond to the reliability of
that output. Thus, we can obtain the stochastic sequence
representing the reliability of each output by taking the
XOR of each output bit stream of the faulty circuit with
the complement of the corresponding output bitstreams
of the fault-free circuit. For a circuit with multiple pri-
mary outputs as is the case here, the stochastic sequence
encoding the joint output reliability can be obtained by
taking the stochastic AND of the outputs of the XOR
gates as the stochastic AND operation on the output of
XOR gates produces a one only if all the corresponding
bits at each XOR gate are one i.e. if all the correspond-
ing bits in the respective outputs of the fault-free and
faulty circuit are same. We then take the complement of
the bit stream at the output of this AND gate to obtain
the stream that encodes vulnerability. This bit stream can
then be decoded to determine the node vulnerability with
the proportion of ones in this stream equivalent to the
vulnerability of the node.
The procedure for computing the vulnerability of a node
described above and depicted in Fig. 5 is summarized as
follows:
Fig. 5 Computation of node vulnerability. Depicts the architecture used to compute the vulnerability of a node. x1 to x7 are the input stochastic bit
streams for each of the seven primary inputs in the Boolean network model. The output bit streams for each of the six output components when
these input sequences are propagated through the circuit with a dysfunctional node (whose vulnerability we want to compute) are denoted by y∗
1
to y∗
6 whereas those for the fault-free circuit are labeled as y1 to y6
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 12 of 59
1. Generate non-Bernoulli streams encoding input
probabilities at each of the primary inputs.
2. Propagate the input binary streams through the
fault-free circuit and obtain a random bit sequence
for each output.
3. Propagate the same input binary streams through the
circuit with a stuck-at fault at the location of the node
whose vulnerability we want to determine and again
obtain a random bit sequence for each output.
4. XOR each primary output sequence from the faulty
circuit obtained in step 3 with the complement of the
corresponding primary output sequence from the
fault-free circuit.
5. AND all the sequences obtained from each XOR gate.
Take the complement of the stream so obtained. The
vulnerability of the node is the fraction of ones in the
resulting bit stream.
Thus, in a nutshell, the node vulnerabilities are obtained
by propagating the initial input stochastic bit streams
encoding the input probabilities through both the faulty
and fault-free circuit, comparing the respective outputs
obtained from each and decoding probabilities from the
resulting streams.
Let
x1, x2, . . . , xN
represent
input
non-Bernoulli
sequences of length L with each sequence represented
as a vector of length L whose ith component is equal
to the ith bit in the sequence. Define the L × N matrix
X = (x⊤
1 x⊤
2 . . . x⊤
N). Thus, each row of this matrix con-
tains the corresponding bits of each of the primary input
streams. The vulnerability vj of node j ∈{1, 2, . . . , P} is
given by:
vj = 1
L
L
k=1
M
i=1
ψi
x = [Xk1, . . . , XkN] , d0, fj
⊕ψ′
i
x = [Xk1, . . . , XkN] , d0, f0
′
(2)
where ′ is the bit-complement operator and ⊕is the binary
XOR operator.
Results and discussion
Simulation results for drug intervention
We use the Boolean network model to determine an appo-
site therapy for each fault. As described in the methods
section, the best targeted therapy for each of the cancer-
inducing faults is the one that under the presence of the
fault, produces the output with the smallest Hamming
weight with the minimal number of drugs. The best ther-
apy for each of the faults is shown in table 1 with the drug
vector defined as before. Note that for certain faults, no
drug vector can improve the output. Such faults are said
to be untestable since no test (drug vector in this case) can
Table 1 Best therapy for each fault
Fault location
Drug vector
1
1000000
2
1000000
3
0100000
4
1000000
5
0011000
6
0011000
7
0000100
8
0001000
9
0001000
10
0000100
11
0000100
12
0000100
13
0000100
14
0000001
15
0000001
16
0010000
17
0010000
18
0000000
19
0000010
20
0000010
21
0000010
22
0000000
23
0000000
24
0000000
rectify the fault. This is because there are no drugs on the
fan-out of these genes. However, all these faults with the
exception of fault 18 are minimally proliferative as they
produce a faulty output with the least possible Hamming
weight of one.
Thus, there are many locations in the gene regulatory
network of prostate cancer where malfunctions can occur
resulting in a cancer that is different, requiring a specific
targeted therapy. The table facilitates arriving at such a
therapy as it maps each malfunction to an appropriate set
of drugs. The look-up table can be used to devise ther-
apies that have a higher likelihood of success since they
are tailored specifically to the molecular abnormalities in
critical pathways and thereby facilitates an individualized
approach to therapy design.
In order to find the best combination therapy across
all possible faults, as discussed in the methods section,
for each drug combination we determine the sum of the
Hamming weights of the output vector across all pos-
sible combinations of faults and choose the drug com-
bination that yields the smallest total. This gives us the
drug cocktail of AZD6244, AZD5363 and Enzalutamide
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 13 of 59
as a combination therapy for advanced prostate cancer.
In a recent study, the drug combination of AZD5363 and
Enzalutamide has demonstrated an impressive response
in prostate cancer models [34]. Moreover, AZD6244 in
partnership with an AKT pathway inhibitor (analogous
to AZD5363), has been proposed as a strategy for the
treatment of CRPC [35]. Thus, we propose that the
aforementioned drug triad which represents a horizon-
tal blockade approach, wherein combination therapy is
used for the concerted pharmacologic inhibition of mul-
tiple compensatory pathways, as a therapeutic modality
that may attenuate prostate cancer survival and growth.
Node vulnerabilities
Using the framework delineated in the methods section,
we quantify the vulnerability of different nodes. The vul-
nerability values obtained are given in Table 2. Vulnera-
bility assessment can be used to identify candidates for
targeted drug development. Nodes whose vulnerabilities
are higher should be presumably better targets for drugs
since potentially therapeutic benefit is more likely for
nodes which are more vulnerable. We observe that the
Table 2 Node vulnerabilities
Node
Vulnerability (%)
1
6.25
2
6.25
3
6.25
4
6.25
5
6.25
6
6.25
7
24.98
8
6.25
9
6.25
10
24.98
11
24.98
12
24.98
13
24.98
14
12.47
15
12.47
16
6.25
17
6.25
18
6.25
19
1.57
20
1.57
21
1.57
22
1.57
23
1.57
24
24.98
AR-mediated signaling axis remains a valid target. Fur-
thermore, we see that dysfunction in the AKT nexus and
the loss of tumor-suppressors have higher vulnerability
values so drugs that attempt to alleviate these aberra-
tions should be efficacious in attenuating tumor growth.
The design of anti-cancer therapeutics directed at the loss
of tumor suppressors has been difficult [36]. Addition-
ally, AKT-selective drug development is challenging due
to its homology with other kinases [37]. These complica-
tions notwithstanding, accelerated development of novel
agents that target these aberrations is warranted. In con-
trast, the vulnerabilities for certain nodes such as those in
the mTOR axis are low indicating that they might not be
attractive targets for drug development. Indeed, marginal
clinical activity has been observed for mTOR inhibition
with agents such as everolimus and temsirolimus failing
to impact tumor proliferation in men with prostate cancer
[4, 38]. Finally, in terms of the key pathways implicated in
the disease we see that castration-resistant prostate can-
cer shows most vulnerability on aggregate to dysfunction
in the AKT pathway. In a study it was demonstrated that
the AKT pathway dominates AR signaling in CRPC [39].
Conclusion
Castration-resistant prostate cancer is a hormone refrac-
tory phenotype of significant morbidity and mortality in
the prostate cancer disease continuum where patients
no longer respond to androgen ablation therapy. The
biomolecular network representing the signaling path-
ways involved in the pathogenesis of this lethal malig-
nancy is translated to a digital circuit. The locations of
possible malfunctions in the digital circuit are identified
and computer simulation of the equivalent model is used
to predict effective therapies that mitigate the effect of
different faults. A prospectively attractive combinatorial
therapeutic strategy for the constellation of abnormalities
is to leverage an AR axis targeted agent in conjunction
with reciprocal inhibitors of other dysregulated pathways
that are fundamental in coordinately driving oncogene-
sis. Proof of principle of clinical use for the proposed
regimen remains to be demonstrated. A reliability (vulner-
ability) analysis methodology of digital circuits premised
on stochastic logic modeling is utilized to quantify the vul-
nerability of the network to the dysfunction in discrete
components in the signaling cascade thereby identifying
key variables as targets for intervention that conceivably
might be exploited by a new generation of novel thera-
peutics. These findings can contribute to the development
of new rational approaches for the possible treatment of
androgen-refractory prostate cancer. There is however a
paucity of companion predictive biomarkers that can be
used for the stratification of patients based on molecular
aberrations in order to prescribe the apposite treatment.
Furthermore, the histological and clinical heterogeneity
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 14 of 59
of CRPC and the inherent redundancy along with the
presence of feedback loops in pathways whose molecu-
lar underpinnings in the context of the disease induction
and development are not yet fully understood, tender
any potential translation into objective clinical efficacy
of therapeutic implications derived from computations
fraught with challenges.
Abbreviations
ADT: Androgen deprivation therapy; AR: Androgen receptor; BCL2: B-cell
lymphoma 2; BN: Boolean network; CDK2: Cyclin dependent kinase 2; CRPC:
Castration resistant prostate cancer; EGF: Epidermal growth factor; GRN: Gene
regulatory network; HBEGF: Heparin binding EGF-like growth factor; IGF:
Insulin-like growth factor; MAPK: Mitogen activated protein kinase; MIMO:
Multi-input multi-output; NRG1: Neuregulin 1; PSA: Prostate specific antigen;
PTEN: Phosphatase and tensin homolog; SP1: Specificity protein 1; TMPRSS2:
Transmembrane protease serine 2
Acknowledgements
Not applicable.
Funding
Publication costs for this article have been funded by the National Science
Foundation (NSF) under grant ECCS-1404314. This work was supported in part
by NSF under grant ECCS-1404314 and the Texas Engineering Experiment
Station (TEES) - Agrilife Center for Bioinformatics and Genomics Systems
Engineering.
Availability of data and materials
Data sharing is not applicable to this article as no datasets were generated or
analyzed during the current study.
Authors’ contributions
OAA developed the computational modeling and simulations and wrote the
manuscript. AD conceived the main idea. Both authors read and approved the
final manuscript.
Competing interests
The authors declare that they have no competing interests.
Consent for publication
Not applicable.
Ethics approval and consent to participate
Not applicable.
About this supplement
This article has been published as part of BMC Bioinformatics Volume 18
Supplement 4, 2017: Selected original research articles from the Third
International Workshop on Computational Network Biology: Modeling,
Analysis, and Control (CNB-MAC 2016): bioinformatics. The full contents of the
supplement are available online at https://bmcbioinformatics.biomedcentral.
com/articles/supplements/volume-18-supplement-4.
Published: 22 March 2017
References
1.
Siegel RL, Miller KD, Jemal A. Cancer statistics 2015. CA: a cancer journal
for clinicians. 2015;65:5–29.
2.
Feng J, Zheng SL, Liu W, Isaacs WB, Xu J. Androgen receptor signaling
in prostate cancer: new twists for an old pathway. J Steroids Hormon Sci.
2011.
3.
Boyd LK, Mao X, Lu YJ. The complexity of prostate cancer: genomic
alterations and heterogeneity. Nat Rev Urol. 2012;9(11):652–64.
4.
Derleth CL, Evan YY. Targeted therapy in the treatment of
castration-resistant prostate cancer. Oncology. 2013;27(7):620–30.
5.
Leo S, Accettura C, Lorusso V. Castration-resistant prostate cancer:
targeted therapies. Chemotherapy. 2010;57(2):115–27.
6.
Chen Y, Clegg NJ, Scher HI. Anti-androgens and androgen-depleting
therapies in prostate cancer: new agents for an established target. Lancet
Oncol. 2009;10:981–91.
7.
Patel JC, Maughan BL, Agarwal AM. Batten AM. Zhang TY: Agarwal N.
Emerging molecularly targeted therapies in castration refractory prostate
cancer. Prostate Cancer; 2013.
8.
Agarwal N, Sonpavde G, Sternberg CN. Novel molecular targets for the
therapy of castration-resistant prostate cancer. Eur Urol. 2012;61(5):
950–60.
9.
Aggarwal R, Ryan CJ. Castration-resistant prostate cancer: targeted
therapies and individualized treatment. Oncologist. 2011;16(3):264–75.
10. Nedumparambathmarath V, Chakrabarti SK, Sreekumar J. Modeling of
gene regulatory networks: a review. J Biomed Sci Eng. 2013;6:223–31.
11. Kaufmann SA. The origins of order: self-organization and selection in
evolution. New York: Oxford University Press; 1993.
12. Helikar T, Konvalina J, Heidel J, Rogers JA. Emergent decision-making in
biological signal transduction networks. Proc Natl Acad Sci. 2008;105(6):
1913–8.
13. Albert R, Thakar J. Boolean modeling: a logic-based dynamic approach
for understanding signaling and regulatory networks and for making
useful predictions. Wiley Interdiscip Rev Syst Biol Med. 2014;6:353–69.
14. Wang R, Saadatpour A, Albert R. Boolean modeling in systems biology:
an overview of methodology and applications. Phys Biol. 2012;9(5).
15. Fauré A, Naldi A, Chaouiya C, Thieffry D. Dynamical analysis of a generic
Boolean model for the control of the mammalian cell cycle.
Bioinformatics. 2006;22(14):e124–e31.
16. Schlatter R, Schmich K, Vizcarra IA, Scheurich P, Sauter T, Borner C,
Ederer M, Merfort I, Sawodny O. ON/OFF and beyond-a Boolean model
of apoptosis. PLoS Comput Biol. 2009;5(12):e1000595.
17. Zhang R, Shah MV, Yang J, Nyland SB, Liu X, Yun JK, Albert R, Loughran
TP. Network model of survival signaling in large granular lymphocyte
leukemia. Proc Natl Acad Sci. 2008;105(42):16308–13.
18. Sridharan S, Varghese R, Venkatraj V, Datta A. Hypoxia stress response
pathways: modeling and targeted therapy. IEEE J Biomed Health Inform.
in press. http://ieeexplore.ieee.org/document/7460895/.
19. Giacomantonio CE, Goodhill GJ. A Boolean model of the gene regulatory
network underlying mammalian cortical area development. PLoS
Comput Biol. 2010;6(9):e1000936.
20. Carver BS, Chapinski C, Wongvipat J, Hieronymus H, Chen Y,
Chandarlapaty S, Arora VK, Le C, Koutcher J, Scher H, Scardino PT,
Rosen N. Reciprocal feedback regulation of PI3K and androgen receptor
signaling in PTEN-deficient prostate cancer. Cancer Cell. 2011;19(5):
575–86.
21. Yuen HF, Abramcyzk O, Montgomery G, Chan KK, Huang YH, Sasazuki T,
Shirasawa S, Gopesh S, Chan KW, Fennell D, Janne P, El-Tanani M,
Murray JT. Impact of oncogenic driver mutations on feedback between
the PI3K and MEK pathways in cancer cells. Biosci Rep. 2012;32(4):413–22.
22. Kanehisa M, Goto S, Furumichi M, Tanabe M, Hirakawa M. KEGG for
representation and analysis of molecular networks involving diseases and
drugs. Nucleic Acids Res. 2010;38:D355–D60.
23. Kanehisa M, Goto S, Hattori M, Aoki-Kinoshita KF, Itoh M, Kawashima S,
Katayama T, Araki M, Hirakawa M. From genomics to chemical genomics:
new developments in KEGG. Nucleic Acids Res. 2006;34:D354–D7.
24. Kanehisa M, Goto S. KEGG: Kyoto Encyclopedia of Genes and Genomes.
Nucleic Acids Res. 2000;28:27–30.
25. Watterson S, Marshall S, Ghazal P. Logic models of pathway biology.
Drug Discov Today. 2008;13(9):447–56.
26. Abramovici M, Breuer MA, Friedman AD. Digital systems testing and
testable design. New York: Wiley-IEEE Press; 1994.
27. Layek R, Datta A, Bittner M, Dougherty E. Cancer therapy design based
on pathway logic. Bioinformatics. 2011;27(4):548–55.
28. Choudhury MR, Mohanram K. Reliability analysis of logic circuits. Comput
Aided Des Integr Circ Syst, IEEE Trans. 2009;28(3):392–405.
29. Han J, Chen H, Liang J, Zhu P, Yang Z, Lombardi F. A stochastic
computational approach for accurate and efficient reliability evaluation.
The Author(s) BMC Bioinformatics 2017, 18(Suppl 4):134
Page 15 of 59
IEEE Trans Comput. 2014;63(6):1336–50.
30. Abdi A, Tahoori MB, Emamian ES. Fault Diagnosis Engineering of Digital
Circuits Can Identify Vulnerable Molecules in Complex Cellular Pathways.
Sci Signal. 2008;1(42):ra10.
31. Zhu P, Aliabadi HM, Uluda˘g H, Han J. Identification of Potential Drug
Targets in Cancer Signaling Pathways using Stochastic Logical Models.
Nat Sci Rep. 2016; 6.
32. Liang J, Han J. Stochastic Boolean networks: an efficient approach to
modeling gene regulatory networks. BMC Syst Biol. 2012; 6.
33. Zhu P, Han J. Asynchronous stochastic Boolean networks as gene
network models. J Comput Biol. 2014;21(10):771–83.
34. Toren P, Kim S, Cordonnier T, Crafter C, Davies BR, Fazli L, Gleave ME,
Zoubeidi A. Combination AZD5363 with enzalutamide significantly
delays enzalutamide-resistant prostate cancer in preclinical models. Eur
Urol. 2015;67(6):986–90.
35. Park H, Kim Y, Sul JW, Jeong IG, Yi HJ, Ahn JB, Kang JS, Yun J, Hwang
JJ, Kim CS. Synergistic anticancer efficacy of MEK inhibition and dual
PI3K/mTOR inhibition in castration-resistant prostate cancer. Prostate.
2015;75(15):1747–59.
36. Dillon LM, Miller TW. Therapeutic targeting of cancers with loss of PTEN
function. Curr Drug Targets. 2014;15:65–79.
37. Bitting RL, Armstrong AJ. Targeting the PI3K/Akt/mTOR pathway in
castration-resistant prostate cancer. Endocr relat Cancer. 2013;20(3):
R83–R99.
38. Sarker D, Reid AHM, Yap TA. de Bono JS. Targeting the PI3K/AKT pathway
for the treatment of prostate cancer. Clin Cancer Res. 2009;15(15):
4799–805.
39. Kaarbø M, Mikkelsen ØL, Malerød L, Qu S, Lobert VH, Akgul G,
Halvorsen T, Mælandsmo GM, Saatcioglu F. PI3K-AKT-mTOR pathway is
dominant over androgen receptor signaling in prostate cancer cells. Anal
Cell Pathol. 2010;32:11–27.
• We accept pre-submission inquiries
• Our selector tool helps you to find the most relevant journal
• We provide round the clock customer support
• Convenient online submission
• Thorough peer review
• Inclusion in PubMed and all major indexing services
• Maximum visibility for your research
Submit your manuscript at
www.biomedcentral.com/submit
Submit your next manuscript to BioMed Central
and we will help you at every step:
|
28361666
|
IGFR1A/B = ( IGF )
RP6SKB1 = ( mTOR AND ( ( ( NOT Temsirolimus ) ) ) ) OR ( PDPK1 ) OR ( ERK1/2 )
p21 = NOT ( ( AKT ) )
mTOR = ( RHEB )
Ras = ( GRB2/SOS )
TMPRSS2 = ( AR/AR AND ( ( ( NOT Enzalutamide ) ) ) )
CDK2-CyclinE = NOT ( ( p21 ) )
Raf = ( PIK3CA AND ( ( ( NOT BKM120 ) ) ) ) OR ( Ras )
AR/AR = ( AKT ) OR ( AR/HSP )
EGFR/ERBB2 = ( EGF )
SRF-ELK1 = ( ERK1/2 AND ( ( ( RP6SKB1 ) ) ) )
MEK1 = ( Raf )
AR/HSP = ( Androgens )
AKT = ( ( NKX3.1 AND ( ( ( NOT PDPK1 AND NOT PTEN ) ) ) ) OR ( PTEN AND ( ( ( NOT PDPK1 AND NOT NKX3.1 ) ) ) ) OR ( PDPK1 ) ) OR NOT ( PDPK1 OR PTEN OR NKX3.1 )
PDPK1 = ( PIP3 )
GRB2/SOS = ( EFGR AND ( ( ( NOT Lapatinib ) ) ) ) OR ( EGFR/ERBB2 AND ( ( ( NOT Lapatinib ) ) ) ) OR ( ERBB2/3 AND ( ( ( NOT Lapatinib ) ) ) ) OR ( IGFR1A/B )
PIK3CA = ( Ras ) OR ( IRS1 ) OR ( ERBB2/3 )
ERK1/2 = ( MEK1 AND ( ( ( NOT AZD6244 ) ) ) )
SP1 = ( ERK1/2 )
ERBB2/3 = ( NRG1 )
PSA = ( AR/AR AND ( ( ( NOT Enzalutamide ) ) ) )
TSC1/2 = NOT ( ( AKT AND ( ( ( NOT AZD5363 ) ) ) ) )
PIP3 = ( ( PIK3CA ) AND NOT ( PTEN ) )
BCL2 = NOT ( ( BAD ) )
RHEB = NOT ( ( TSC1/2 ) )
EFGR = ( HBEGF ) OR ( EGF )
IRS1 = ( IGFR1A/B AND ( ( ( NOT Cixutumumab ) ) ) )
BAD = NOT ( ( AKT ) OR ( RP6SKB1 ) )
|
Logical modeling of lymphoid and myeloid cell
specification and transdifferentiation
Samuel Collombeta,1, Chris van Oevelenb,2, Jose Luis Sardina Ortegab,2, Wassim Abou-Jaoudéa, Bruno Di Stefanob,3,
Morgane Thomas-Cholliera, Thomas Grafb,c,1, and Denis Thieffrya,1
aComputational Systems Biology Team, Institut de Biologie de l’Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Ecole Normale Supérieure, Paris
Sciences et Lettres Research University, 75005 Paris, France; bHematopoietic Stem Cells, Transdifferentiation, and Reprogramming Team, Gene Regulation,
Stem Cells, and Cancer Program, Center for Genomic Regulation, Barcelona Institute for Biotechnology, 08003 Barcelona, Spain; and cUniversitat Pompeu
Fabra, 08002 Barcelona, Spain
Edited by Ellen V. Rothenberg, California Institute of Technology, Pasadena, CA, and accepted by Editorial Board Member Neil H. Shubin November 18, 2016
(received for review September 1, 2016)
Blood cells are derived from a common set of hematopoietic stem
cells, which differentiate into more specific progenitors of the myeloid
and lymphoid lineages, ultimately leading to differentiated cells. This
developmental process is controlled by a complex regulatory network
involving cytokines and their receptors, transcription factors, and
chromatin remodelers. Using public data and data from our own mo-
lecular genetic experiments (quantitative PCR, Western blot, EMSA) or
genome-wide assays (RNA-sequencing, ChIP-sequencing), we have
assembled a comprehensive regulatory network encompassing the
main transcription factors and signaling components involved in my-
eloid and lymphoid development. Focusing on B-cell and macrophage
development, we defined a qualitative dynamical model recapitulat-
ing cytokine-induced differentiation of common progenitors, the ef-
fect of various reported gene knockdowns, and the reprogramming
of pre-B cells into macrophages induced by the ectopic expression of
specific transcription factors. The resulting network model can be
used as a template for the integration of new hematopoietic differ-
entiation and transdifferentiation data to foster our understanding
of lymphoid/myeloid cell-fate decisions.
gene network | dynamical modeling | hematopoiesis | cell fate |
cell reprogramming
H
ematopoiesis is the process through which all blood cells are
produced and renewed, starting from a common population
of hematopoietic stem cells (HSCs) (1). HSCs differentiate into
lineage-specific progenitors with restricted differentiation potential
and expressing specific surface markers (Fig. 1A). Loss- or gain-of-
function experiments targeting transcription factors (TFs) or
signaling components have led to the identification of factors re-
quired for specific developmental steps. Some factors are required
for the development of entire lineages (e.g., Ikaros for lymphoid
cells), whereas others are needed only at late stages of cell-type
specification (e.g., the requirement for the paired-box factor Pax5
after the pro–B-cell stage). These factors cross-regulate each other to
activate one gene-expression program and silence alternative ones.
Although cell commitment to a specific lineage was long con-
sidered irreversible, recent reprogramming experiments emphasized
the pervasive plasticity of cellular states. Indeed, the ectopic ex-
pression of various regulatory factors (mainly TFs and signaling
components) can enforce the establishment of new gene-expression
programs in many kinds of differentiated cells (2). Strikingly, pluri-
potency can be induced in somatic cells by forcing the expression of
a handful of TFs, enabling further differentiation into any cell type
(3). In the hematopoietic system, TF-induced transdifferentiation
between erythroid and myeloid cells and between lymphoid and
myeloid cells has been described (4).
In this study, we focus on B-cell and macrophage specification
from multipotent progenitors (MPs) and on TF-induced trans-
differentiation between these lineages. Ectopic expression of the
myeloid TF C/EBPα (CCAAT/enhancer-binding protein alpha,
encoded by the Cebpa gene) can induce B cells to transdifferentiate
into macrophages (Fig. 1A, red arrows) (5). C/EBPα is also required
for the transition from common myeloid progenitors (CMPs) to
granulocyte-macrophage progenitors (GMPs), and mutation in
this gene can result in acute myeloid leukemia (6). Understanding
the molecular mechanisms by which such factors can induce cell-
fate decisions is of primary importance and might help in the
development of novel therapeutic strategies.
Computational modeling of regulatory networks is increasingly
recognized as a valuable approach to study cell-fate decisions. In-
deed, the integration of the available information about gene
regulation into a common formal framework allows us to identify
gaps in our current knowledge, as successfully shown in previous
studies on the differentiation of hematopoietic cells (7). Dynamic
analysis can reveal nontrivial properties, including transient phe-
nomena, and can be used to identify key regulatory factors or in-
teractions involved in the control of cell-fate commitment (8, 9).
Furthermore, genome-wide approaches such as ChIP-sequencing
(ChIP-seq) can unveil novel regulations to be further incorporated
in a gene-network model (10). Here, we combined a logical mul-
tilevel formalism, capturing the main qualitative aspects of the
dynamics of a regulatory network in the absence of quantitative
kinetic data (11), with a meta-analysis of all available ChIP-seq
datasets for a selection of TFs, revealing tens of previously un-
known regulations. We then performed iterations of computational
simulations, followed by comparisons with experimental data and
adjustments of the model, to identify caveats in our model and to
test the effect of putative regulations in silico before confirming
them experimentally (Fig. 1B).
This paper results from the Arthur M. Sackler Colloquium of the National Academy of
Sciences, “Gene Regulatory Networks and Network Models in Development and Evolu-
tion,” held April 12–14, 2016, at the Arnold and Mabel Beckman Center of the National
Academies of Sciences and Engineering in Irvine, CA. The complete program and video
recordings of most presentations are available on the NAS website at www.nasonline.org/
Gene_Regulatory_Networks.
Author contributions: S.C., C.v.O., J.L.S.O., T.G., and D.T. designed research; S.C., C.v.O.,
J.L.S.O., and B.D.S. performed research; S.C., W.A.-J., B.D.S., M.T.-C., T.G., and D.T.
analyzed data; and S.C., J.L.S.O., W.A.-J., M.T.-C., T.G., and D.T. wrote the paper.
The authors declare no conflict of interest.
This article is a PNAS Direct Submission. E.V.R. is a guest editor invited by the Editorial
Board.
Data deposition: The ChIP-seq data for EBF1 and Foxo1 in pre-B cell lines and during trans-
differentiation have been deposited in the Gene Expression Omnibus (GEO) database (ac-
cession code GSE86420). The final model has been deposited in the BioModels database
(accession no. 1610240000).
1To whom correspondence may be addressed. Email: denis.thieffry@ens.fr, samuel.
collombet@ens.fr, or thomas.graf@crg.eu.
2C.v.O. and J.L.S.O. contributed equally to this study.
3Present addresses: Department of Molecular Biology, Center for Regenerative Medicine
and Cancer Center, Massachusetts General Hospital, Boston, MA 02114; Department of
Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138; Harvard
Stem Cell Institute, Cambridge, MA 02138; and Harvard Medical School, Cambridge,
MA 02138.
This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
1073/pnas.1610622114/-/DCSupplemental.
5792–5799
|
PNAS
|
June 6, 2017
|
vol. 114
|
no. 23
www.pnas.org/cgi/doi/10.1073/pnas.1610622114
Results
Gene Network Controlling B-Cell and Macrophage Specification. To
build a model of the gene-regulatory network controlling B-cell and
macrophage specification from common progenitors, we first per-
formed an extensive analysis of the literature to identify the TFs
and signaling pathways controlling these events. The TF PU.1
(encoded by the Spi1 gene) is required for the normal development
of both lymphoid and myeloid cells (12). The development of
common lymphoid progenitors (CLPs) depends on the TFs Ikaros
(encoded by Ikzf1) and E2a (encoded by the transcription factor 3
gene Tcf3) (Fig. 1A) (13, 14). The B-cell lineage is further con-
trolled by Mef2c, the interleukine 7 receptor (IL7r), Ets1, Foxo1,
Ebf1, and Pax5 (15, 16, 17). The specification of the myeloid GMPs
depends on C/EBPα (6), which is regulated by Runx1 (runt-related
transcription factor 1) (18). Macrophage specification further relies
on the macrophage colony-stimulating factor (M-CSF) receptor
(CSF1r), on the up-regulation of PU.1, and on Cebpb and the Id
proteins (including Id2) (19, 20). The TFs Egr and Gfi1 repress each
other to specify macrophage versus granulocyte lineages (21); Gfi1
also is important for B-cell differentiation (22).
Finally, to distinguish among the different cell types, we further
consider the B-cell marker CD19, the macrophage marker Mac1
(also called “Cd11b,” encoded by the Itgam gene), and the cytokine
receptor Flt3, which is expressed specifically on MPs and CLPs.
We then carried out an extensive review of the literature to
collect information about cross-regulations between the selected
factors and grouped these regulations into four classes, depending
on the available evidence: (i) functional effect, e.g., an effect
inferred from gain- or loss-of-function experiments (which could be
either direct or indirect); (ii) physical interaction, e.g., TF binding
at a promoter or enhancer; (iii) physical and functional evidence,
suggesting a direct regulation; and (iv) fully proven regulation, e.g.,
evidence of functional effect and physical interaction along with
reported binding-site mutations affecting the functional effect or
reporter assays demonstrating cis-regulatory activity. Altogether,
we gathered a total of 150 items of experimental evidence (Dataset
S1) supporting 79 potential regulations (Fig. S1A).
Many of these regulations are sustained only by functional
evidence. To assess whether they could correspond to direct
regulations, we analyzed public ChIP-seq datasets targeting each of
the TFs considered in our network, amounting to 43 datasets for 10
TFs in total (Dataset S2). We systematically looked for peaks in the
“gene domain” (23) coding for each component involved in the
network (Materials and Methods). This ChIP-seq meta-analysis
confirmed 26 direct regulations (Fig. 2A, green or red cells with a
star) and pointed toward 66 additional potential transcriptional
regulations (gray cells with a star). For example, at the Spi1 locus,
we confirmed the binding of Ikaros at known enhancers, where it
was previously reported to limit the expression of Spi1 together
with a putative corepressor (24). Because we also found that Pax5,
Ebf1, and Foxo1 bind to the same sites (Fig. 2B), we suggest that
these factors could act as corepressors. Ectopic expression of
Foxo1 in macrophages induced a reduction of Spi1 expression
(Fig. S1B), further confirming this negative regulation.
C/EBPα Directly Represses B-Cell Genes. We have previously reported
that C/EBPα can enforce B-cell TF silencing by increasing the
expression of the histone demethylase Lsd1 (Kdm1a) and the his-
tone deacetylase Hdac1 at the protein level and that these enzymes
are required for the decommissioning of B-cell enhancers and the
silencing of the B-cell program (25). Because key B-cell regulators
such as Foxo1, Ebf1, and Pax5 are repressed after 3 h of C/EBPα
induction (Fig. S1C), we wondered whether C/EBPα could be di-
rectly responsible for this early effect. To verify this hypothesis, we
reanalyzed data from ChIP-seq for C/EBPα after 3 h of induction
in a reprogrammable cell line (26). As expected, we detected
binding of C/EBPα at the cis-regulatory elements of Foxo1 (Fig.
2C), Ebf1, Pax5, IL7r, and Mef2c genes (Fig. S1C), supporting their
direct repression by C/EBPα.
Furthermore, C/EBPβ also can induce transdifferentiation of pre-
B cells (5), and it has been shown that C/EBPβ can rescue the
formation of granulocytes in C/EBPα-deficient mice (27). Moreover,
C/EBPβ almost always binds at C/EBPα-binding sites (Fig. 2A), as
exemplified by the Spi1 locus (Fig. 2B). These findings suggest a
A
B
literature
Fig. 1.
(A) Schematic representation of hematopoietic cell specification. Genes in red are required for progression at the corresponding steps. C/EBPα-induced
transdifferentiation is indicated by red arrows from B-lineage cells to macrophages. (B) Iterative modeling workflow. A model is first built based on the literature
and is used to predict dynamical behaviors (cell phenotype, differentiation, reprogramming, and so forth). Predictions then are compared with experimental data;
when the predictions and experimental data agree, further predictive simulations are performed; when they do not agree, further regulations are inferred from
ChIP-seq data and are integrated into the model until simulations fully agree with data.
Collombet et al.
PNAS
|
June 6, 2017
|
vol. 114
|
no. 23
|
5793
SYSTEMS BIOLOGY
COLLOQUIUM
PAPER
redundancy between these two factors in the regulation of their
target genes (at least in those considered here), and we integrated
this redundancy in our model.
Dynamical Modeling Using Multilevel Logic. The core components
and regulations collected from our analysis of the literature and
ChIP-seq datasets were assembled in a regulatory graph using the
GINsim software (Fig. 3).
Validating all the predicted regulations (Fig. 2A, gray cells with a
star) experimentally would be a daunting task. Instead, we focused
on a selection of these regulations (depicted by the gray arrows in
Fig. 3) and used dynamical modeling to assess their impact on
cell specification.
To transform our regulatory graph into a predictive dynamical
model, we took advantage of a sophisticated logical (multilevel)
formalism. More precisely, we associated a discrete variable with
each regulatory component. These variables usually take two values
(0 or 1) but can be assigned more values whenever justified.
Regulations are combined into logical rules using the Boolean op-
erators NOT, AND, and OR, to define the conditions enabling the
activation of each model component (Materials and Methods). This
formalism relies essentially on qualitative information and allows the
simulation of relatively large network models (encompassing up to a
few hundred components). It should be noted that the value 0 does
not necessarily imply that a factor is not expressed at all but rather
that its level is insufficient to affect its targets significantly. PU.1
is the only factor for which we found clear evidence supporting
a distinction between two functional (non-0) levels (21). Conse-
quently, we assigned a ternary variable (taking the values 0, 1, or 2)
to this node and assigned Boolean variables (i.e., taking the values
0 or 1) to the other nodes.
Regarding the definition of the logical rules, we first considered
the regulations supported by both functional and physical evidence
(depicted as green and red arrows in Fig. 3). As a default, we re-
quired that all activators but no inhibitor to be present to enable
target activation and further adjusted the rules based on in-
formation gathered from the literature (see the rules in Materials
and Methods and Dataset S3). As mentioned before, we then added
selected regulations inferred from our ChIP-seq meta-analysis
(depicted as gray arrows in Fig. 3) to refine our model.
Modeling Different Cell-Type Phenotypes. We first assessed whether
our model properly accounts for progenitor, B-cell, and macrophage
gene-expression patterns. Because stable states capture the long-
term behavior associated with the acquisition of gene-expression
patterns during cell specification, we computed all the stable states
of our model using GINsim software (28) and compared them with
gene-expression data (Fig. 4A) (29). We initially found that our
stable states were largely inconsistent with known patterns of gene
expression (Fig. S2A), revealing important caveats in the published
data on which we based our model.
A first caveat concerned the regulation of Cebpa. Indeed, Cebpa
is not expressed in lymphoid cells, although its well-known activa-
tors PU.1 (Spi1) and Runx1 are expressed in both B cells and
Foxo1
Ebf1
Bcell
Pax5
Bcell
Spi1
Bcell
Ikzf1
Bcell
Cebpa
B+Cebpa
Cebpa
GMP
Cebpa
Mac
Cebpb
Mac
H3K27ac
H3K27ac
Spi1
Ebf1
Bcell
Pax5
Bcell
Spi1
Bcell
Ikzf1
Bcell
Cebpa
GMP
Cebpa
Mac
Cebpb
Mac
Pu1
Mac
Runx1
Mac
Gfi1
MP
Runx1
MP
Foxo1
Bcell
Causal
+
-
+/-
Cebpa
Cebpb
Regulator
Target
Functional
Func+Phy
Physical
Cd19
Pax5
Ebf1
Foxo1
Tcf3
Il7r
Ets1
Mef2c
Flt3
Ikzf1
Gfi1
Runx1
Spi1
Csf1r
Cebpa
Cebpb
Egr2
Id2
Itgam
Cd19
Pax5
Ebf1
Foxo1
Tcf3
Il7r
Ets1
Mef2c
Flt3
Ikzf1
Gfi1
Runx1
Csf1r
Id2
Itgam
*
*
*
*
*
*
*
*
*
*
*
*
*
* *
*
*
*
*
*
*
*
* *
* * *
*
*
*
*
*
*
*
*
*
*
*
*
*
*
*
*
*
Spi1
Egr2
*
*
*
*
*
*
*
*
*
*
* *
*
* *
* *
* *
*
*
*
*
*
* *
* *
*
*
*
*
* *
* *
*
* *
* *
* *
* *
*
*
*
Data from ChIP-seq
A
B
C
10kb
100kb
*
Fig. 2.
(A) Heatmap showing the regulations inferred from the literature and
from ChIP-seq meta-analysis. (B) ChIP-seq signals and peaks (under signal) at
the Spi1 locus. Black frames indicate known enhancers (24). The vertical axes
represent reads per million (RPM) (maximum: 2 RPM for Ebf1 and Ikaros, 1.5
RPM for Foxo1, 1 RPM for Runx1 and Gfi1, 5 RPM for other TF). (C) ChIP-seq
signals and peaks (under signal) at the Foxo1 locus. Black frames indicate B-cell
enhancers in which C/EBPα binding is detected. The vertical axes represent
RPM (maximum: 2 RPM for Ebf1, 5 RPM for other TFs, 3 RPM for H3K27ac).
Note that Pax5 and Ikaros peaks are located downstream of the first exon and
all other peaks are upstream of the TSS.
Mac1
Id2
Egr1
Cebpb
Cebpa
Csf1r
Runx1
Gfi1
Ikzf1
Flt3
Mef2c
Ets1
Il7r
E2A
Foxo1
Ebf1
Pax5
Cd19
Csf1
Spi1
Il7
Csf1r
activated
Il7r
activated
Fig. 3.
A regulatory graph depicting the interactions inferred from the litera-
ture and ChIP-seq meta-analyses. Nodes represent genes (except for CSF1r_act
and Il7r_act, which represent the activated forms of cytokine receptors), and
arrows denote regulatory interactions. Orange nodes represent factors expressed
in macrophages, purple nodes represent factors expressed in progenitors, and
blue nodes represent factors expressed in B-lineage cells. Ellipses represent
Boolean components; the rectangle emphasizes the ternary component Spi1.
Green and red edges correspond to activations and inhibitions, respectively. Gray
edges denote the regulations predicted by the ChIP-seq meta-analysis, which
were included in the model to increase consistency with expression data.
5794
|
www.pnas.org/cgi/doi/10.1073/pnas.1610622114
Collombet et al.
macrophages. Therefore, our model exhibited only Cebpa+ stable
states (Fig. S2A), suggesting that an inhibitory regulation of Cebpa
was missing during lymphoid specification. Foxo1, a factor control-
ling the early steps of B-cell commitment (30), stands as a relevant
candidate. To test this hypothesis, we performed ChIP-seq for
Foxo1 in our pre–B-cell line and observed binding at the Cebpa
promoter, suggesting a physical interaction and potential direct
regulation of Cebpa (Fig. S2B). To test if Foxo1 has a functional effect
on Cebpa expression, we ectopically expressed it in a macrophage cell
line (RAW) and found a significant down-regulation of Cebpa (Fig.
S2C), suggesting a direct negative regulation of Cebpa by Foxo1.
We therefore refined our initial model by including this additional
regulation (see the rule associated with Cebpa in Dataset S3).
A second caveat revealed by our model analysis concerned the
regulation of Tcf3 (encoding E2a). Indeed, E2a was expressed in all
the stable states, even after Cebpa repression by Foxo1 was in-
cluded (Fig. S2D), although E2a has been shown to be expressed in
MPs and in lymphoid cells but not in myeloid cells. Moreover, the
only factor in our model expressed in MPs and regulating E2a is
PU.1, which is also known to be expressed in myeloid cells, thus
suggesting a missing regulation of E2a. However, despite our ef-
forts, we could not find any evidence for a myeloid repressor of
E2a in either the literature or our ChIP-seq data meta-analysis.
Turning to putative activators of E2a, we focused on Ikaros. In-
deed, like E2a, Ikaros is required for lymphoid development, and
its knockout entails a loss of lymphocytes similar to that seen with
E2a knockout. Interestingly, we found that Ikaros binds the E2a
promoter in B cells (Fig. S2E), suggesting a direct activation of E2a
by Ikaros. Hence, we further refined our model by including this
regulation (see the rule associated with E2a in Dataset S3).
More surprising was the high expression of Egr2 observed in pro/
pre-B cells. We also found expression of the related factor Egr1 in
two different datasets (Fig. S2 F and G). It has been reported that
Egr1/2 cross-inhibits Gfi1, the first favoring macrophage specifi-
cation and the second favoring B lineage (21). However, although
this study shows that Egr2 has an effect on the differentiation
potential of MPs, it does not demonstrate that this factor is indeed
not expressed in B cells or that it can antagonize the expression of
B-cell genes. To assess the expression of Egr2, Egr1, and Gfi1 at
the protein level, we performed Western blots for these proteins in
B cells and macrophages. We were able to detect all three proteins
in B cells (Fig. S2H), confirming the gene-expression data. We
therefore propose that some late B-cell factors activate both Gfi1
and Egr2, overcoming their cross-inhibitions. Because Pax5 was the
only B-cell factor found in our meta-analysis to bind to Gfi1 and
Egr2 loci (Fig. 2A), we consider it to be an activator of both Gfi1
and Egr2 (see corresponding rules in Dataset S3).
When analyzing the resulting refined model, we found that its
stable states correspond well to CLPs, GMPs, B-lineage cells, and
macrophages, as defined by the known patterns of gene expression
(Fig. 4B). For some genes, we obtained apparent discrepancies
between expression data and stable state values; these discrep-
ancies can be attributed to model discretization (see SI Materials
and Methods for more details).
Our analysis points to previously unrecognized regulators of E2a
and Cebpa that are important at the onset of lymphoid and myeloid
specification and introduces refinements of the regulations of Egr2
and Gfi1. After incorporating these regulations in our model, we
used it to study the dynamics of B-cell and macrophage specification.
Specification of B-Cell and Macrophage Precursors from MPs. To improve
our understanding of the transcriptional regulation of hematopoietic
cell specification, we performed several iterations of hypothesis-
driven simulations and comparisons with experimental data, fol-
lowed by model modifications to solve remaining discrepancies.
First, using GINsim software, we simulated the specification of
MPs, defined by the expression of Spi1, Runx1, Ikzf1, Gfi1, and Flt3.
In the absence of environmental signals, we found that our model
can lead to two different stable states corresponding to GMPs and
CLPs (Fig. 5A). Upon stimulation with both CSF1 and IL7, the
system tends to two new stable states, corresponding to macro-
phages and B lineage cells, respectively. These simulations thus re-
capitulate the commitment of cells to GMP- and CLP-associated
states and their loss of potential for alternative lineages.
Next, using stochastic simulations (see Materials and Methods and
ref. 31 for more details), we analyzed the evolution of the fraction of
cells expressing distinct factors associated with specific cell lineages
starting with the same initial state (MPs) and environmental con-
ditions (initially no stimulation, followed by stimulation with Csf1
and Il7). Our results show two waves of gene activation for both
myeloid and lymphoid factors. The first wave corresponds to the
progenitor (GMP or CLP) expression programs, and the second one
corresponds to terminally differentiated cells (macrophages or B
cells) (Fig. 5B, Top and Middle). The evolution of the different cell
populations (defined by the gene-expression signatures indicated in
Dataset S4) was consistent with our logical simulations, with a rapid
decrease of the MP population followed by the specification toward
GMPs and CLPs and then by their differentiation into macrophages
and B cells, respectively (Fig. 5B, Bottom). The proportions of
myeloid and lymphoid cells were ∼75 and 25%, respectively, in
qualitative agreement with the higher proportion of myeloid cells
present in the bone marrow (32). Tentatively, this asymmetry
could be encoded in the regulatory circuitry rather than merely
being the result of differences in proliferation rates. A sensitivity
analysis further revealed that the proportion of lymphoid and
myeloid cells was affected only by changes in the up-regulation
rates of Cebpa, Foxo1, and E2a (Fig. S2I), supporting the key
function of Cebpa and Foxo1 in the commitment decision (E2a
being required for Foxo1 expression).
To obtain more comprehensive insights into the alternative
trajectories underlying myeloid and lymphoid lineage specification,
we clustered the logical states (Fig. 5A) to generate a hierarchical
(acyclic) graph (28) in which all the states with a similar potential
(i.e., leading to the same attractors or differentiated states) are
Fig. 4.
(A) Gene-expression values (microarrays) in lymphoid/myeloid pro-
genitors (LLPP), B cells, and macrophages (Mac) (29). These values are relative to
the highest expression value. (B) Context-dependent stable states computed for
the model. A yellow cell denotes the inactivation of the corresponding com-
ponent, a red cell represents maximal activation (1 for Boolean components, 2
for Spi1), and an orange cell represents an intermediate level (1) for Spi1.
Collombet et al.
PNAS
|
June 6, 2017
|
vol. 114
|
no. 23
|
5795
SYSTEMS BIOLOGY
COLLOQUIUM
PAPER
clustered in a single node (Fig. 5C). Interestingly, this analysis
shows that the cell decision between GMPs and CLPs depends
mainly on the concurrent activation of Cebpa and Foxo1, em-
phasizing the importance of these factors in early hematopoietic
progenitor specification.
Simulation of Documented Genetic Perturbations. Next, we simulated
the effects of well-documented gene loss-of-function experiments on
progenitor cell specification. Our simulations faithfully recapitulated
the effects of various published gene-ablation experiments (Dataset
S5). For example, Cebpa knockout in MPs results in the loss of the
stable states associated with GMPs and macrophages (Fig. 5D), in
agreement with the reported impact in vivo (33). Pax5 knockout
does not affect the formation of the progenitors but blocks the de-
velopment of the B-cell lineage at the pro-B stage and prevents the
acquisition of the terminal B-cell marker Cd19 (Fig. 5E), in agree-
ment with published experimental data (34).
However, the simulation of Spi1 knockout does not reproduce
the reported viability of B cells in Spi1-knockout mice (35). This
discrepancy arose because, in our model, Spi1 is required for the
expression of the B-cell factors E2a, Ebf1, and Il7r. Introducing
additional cross-activations between the B-cell factors and releasing
the requirement of Runx1 for Ebf1 up-regulation and of Mef2c for
Il7r activation could rescue the expression of the B-cell factors.
When we refined the corresponding rules accordingly (Dataset S3),
the resulting model showed a stable state corresponding to B-cell
patterns in the Spi1-knockout condition. However, such patterns
cannot be reached from a Spi1−/−MP state, because the cells end
up with a complete collapse of gene expression (Fig. 5F).
Dynamical Analysis of Transdifferentiation. Next, we analyzed in
silico the transdifferentiation of pre-B cells into macrophages upon
C/EBPα induction. We first simulated the behavior of B cells under
a permanent induction of C/EBPα in the presence of CSF1 and
IL7. The system converged toward a single stable state corre-
sponding to macrophages, which does not further require induction
of exogenous C/EBPα (Fig. S3A), in accordance with published
reports (5).
We then focused on the effect of transient inductions of C/EBPα.
We have previously shown with our β-estradiol–inducible pre–B-cell
A
C
D
B
E
F
environmental
Fig. 5.
(A) State transition graph generated by simulating the model starting from the unstable MP state in the absence of cytokine (Upper) and after the
addition of CSF1 and IL7 (Lower Left and Lower Right). Nodes denote states, and arrows represent transitions between states. (B) Stochastic simulations
showing the evolution over time, before and after cytokine exposition, of the fractions of cells expressing specific macrophage factors (Top), B-cell factors
(Middle), and cell-type signatures (Bottom). The x and y axes represent time (in arbitrary units) and fractions of positive cells, respectively. (C) Hierarchical
transition graph corresponding to the state transition graph in A. Nodes represent clusters of states, and arrows denote the possible transitions between the
clusters. The labels associated with the edges highlight the crucial transitions involved in the decision between B-cell and macrophage specifications. (D–F)
Schematic representations and stochastic simulations of the effects of Cebpa knockout (D), Pax5 knockout (E), or Spi1 knockout (F) on the differentiation of
MPs, compared with the wild-type situation in A and B. In the cartoons, the wild-type stable states (cell types) and transitions that are lost in each mutant are
displayed using light gray arrows and shading. MP, B cells, and macrophages are represented in purple, blue, and red, respectively.
5796
|
www.pnas.org/cgi/doi/10.1073/pnas.1610622114
Collombet et al.
line that a 24-h induction of C/EBPα followed by washout of the
inducer was sufficient to trigger irreversible reprogramming (36).
Shorter inducer exposure times led to the formation of two pop-
ulations: one converting into macrophages, and the other initiating
transdifferentiation but returning to a B-cell state. A simulation of
this process testing all possible pulse durations at once (Materials
and Methods) confirms that, depending on the duration of C/EBPα
induction, B cells can be reprogrammed to macrophages or can go
back to a B-cell state (the state transition graph for such simulations
cannot be displayed because it contains more than 30,000 states).
Aiming at identifying the commitment point of reprogramming,
we further analyzed the resulting hierarchical transition graph (Fig.
6A). Because endogenous Cebpa becomes activated very late
during transdifferentiation (at about 48 h; see Fig. S3B), notably
after the commitment point (∼24 h), we focused on the Cebpa−
states (i.e., with Cebpa = 0) leading to the sole macrophage stable
state (Fig. 6A, Lower). Some of these states expressed Foxo1,
suggesting that the inhibition of Cebpa by Foxo1 can be overcome,
in contrast with what happens during the specification of GMPs
and CLPs from MPs (Fig. 5C). Interestingly, we found that these
Cebpa−states show low constraints on B-cell factors, because only
Pax5 must be down-regulated. Furthermore, all Cebpa−states
expressed Cebpb and Spi1 at a high level, whereas Pax5 was the
only B-cell factor required to be inactivated. Finally, some states
were found to be Csf1r−, but only when Gfi1 is silenced (along with
its activator Ikaros, at least when its repressor Egr2 is not
expressed), because Gfi1 can block high Spi1 expression (21).
Turning to stochastic simulations, we observed the expected loss
of B-cell and gain of macrophage phenotypes for both permanent
and transient C/EBPα-induced expression (Fig. S3C). However,
these more quantitative simulations also revealed some inconsis-
tencies. (i) Cebpa is reactivated very rapidly; this discrepancy can be
circumvented by lowering the kinetic rate of Cebpa up-regulation.
(ii) The timing of the repression of B-cell genes and that of the loss
of CD19 marker roughly coincide; however, we observed that B-cell
genes are transcriptionally repressed very rapidly (after 3 h; see Fig.
S1C), whereas CD19 protein is lost only after 24 h (36). (iii) Our
model also does not properly capture the fact that short C/EBPα
pulses result in the loss of CD19+ cells, which are regained after
Cebpa inactivation (36), suggesting that reversion of reprogram-
ming is possible after short induction.
The last two points suggest that B-cell TFs are rapidly down-
regulated at the transcriptional level but that the corresponding
proteins are retained in transdifferentiating cells for longer times,
facilitating reversion of the reprogramming. To address this possi-
bility, we performed a ChIP-seq for Ebf1 at several time points upon
permanent induction of Cebpa. Indeed, although Ebf1 RNA de-
creased by 50% after 3 h of C/EBPα induction (Fig. S1C), we ob-
served that Ebf1 binding was lost only after 24 h of induction (Fig.
6B). We therefore added a delay in B-cell factor protein degrada-
tion to our model (Materials and Methods), resulting in a better fit
with the observed timing of events during transdifferentiation for
both permanent and transient C/EBPα induction (Fig. 6C).
In conclusion, our analysis suggests an important role for the
Egr2-Gfi1-PU.1– and C/EBPβ-PU.1–positive loops in the irre-
versible commitment during transdifferentiation and emphasizes
the importance of the balance between protein degradation and
transcriptional regulation kinetics in the reversibility of the
reprogramming.
Simulations of Combined Perturbations During Transdifferentiation.
Finally, we analyzed the effects of various TF gain-/loss-of-functions
on Cebpa-induced reprogramming, combining C/EBPα induction
with a knockdown of Spi1 or Cebpb or with a constitutive ex-
pression of E2a, Ebf1, Pax5, Foxo1, or Gfi1 (Fig. 7). As pre-
viously shown (26), only the Spi1 knockdown is able to block
Ebf1(protein)
E2a
Foxo1
Ebf1 (gene)
B cell
Mac
Ikzf1
Pax5
Cd19
1
0.75
0.50
0.25
0
1
0.75
0.50
0.25
0
1
0.75
0.50
0.25
0
+Cebpa
A
0h
3h
12h
24h
0h
18h
Ebf1
H3K27ac
B cell
Mac
B cell
Mac
1
0.75
0.50
0.25
0
Fraction of positive cells
Time
Bcell
Bcell
Mac
basin of
attraction
basin of
attraction
+C/EBPa
6540
states
938
states
24
states
B
Cebpa negative states
Id2
Egr2
Cebpb
Cebpa
Csf1r
Spi1
Runx1
Gfi1
Ikzf1
Mef2c
Ets1
Il7ra
Tcf3
Foxo1
Ebf1
Pax5
Variable state
C
0
1
0 or 1
5kb
0h
3h
12h
24h
0h
18h
Ebf1
H3K27ac
0h
3h
12h
24h
0h
18h
Ebf1
H3K27ac
20kb
10kb
Fig. 6.
(A, Upper) Hierarchical transition graph of the simulation of B-cell transdifferentiation upon transient C/EBPα expression, taking into account all
possible C/EBPα pulse durations. Nodes represent clusters of states, and arcs correspond to transitions between these clusters. (Lower) Cebpa−states (rows) of
the basin of attraction of the macrophage stable state. (B) ChIP-seq signals and peaks (under signal) in B cells (in blue, time point 0 h) and after induction of
C/EBPα (at 3, 12, and 24 h). The vertical axes represent RPM (maximum, 5 RPM). (C) Stochastic simulations of the fraction of cells expressing different B-cell
factors (Top Panel) and cell-population signatures (Lower Three Panels) during transdifferentiation upon permanent (Upper Two Panels) or transient (Lower
Two Panels) C/EBPα ectopic expression. The corresponding induction durations (in arbitrary units) are indicated by the black lines above each panel.
Collombet et al.
PNAS
|
June 6, 2017
|
vol. 114
|
no. 23
|
5797
SYSTEMS BIOLOGY
COLLOQUIUM
PAPER
transdifferentiation fully under permanent induction of C/EBPα.
The analysis of the HTG obtained for transient C/EBPα induction
in Spi1−cells indicates that, when Ebf1 is inhibited, the expression
of all genes collapses (Fig. S3D). Cebpb knockdown does not
block pulse-induced transdifferentiation, but then all committed
cell states become Cebpa+, suggesting that Cebpb knockdown
could impair commitment if C/EBPα induction is stopped before
the reactivation of endogenous Cebpa; this hypothesis remains
to be tested. Interestingly, the simulation of the constitutive
expression of both Foxo1 and Pax5 results in states expressing a
mixture of myeloid and lymphoid genes, pointing toward ab-
errant reprogramming (Fig. S3E).
Discussion
Models of regulatory networks are classically built from detailed
reviews of the literature. Despite the massive use of high-
throughput assays in the last decade, taking advantage of such data
for the construction of new models or to improve preexisting ones
remains challenging. Here we combined a meta-analysis of ChIP-
seq data with a dynamical model analysis to uncover important
regulations. Such a meta-analysis requires an extensive manual
curation of the datasets.
It would be tempting to explore the different logical rules in a
more unsupervised way by building all possible models with all
combinations of regulations and testing their accuracy in silico.
Although this approach has been used previously (37), it can be
applied only to a subset of possible combinations (e.g., testing the
addition or removal of regulations under a general logical rule,
such as requiring all activators but none of the inhibitors to enable
the activation of a component) and impose certain technical
constraints (e.g., limitation to Boolean variables or to synchronous
updating). In this study, we first built a model based on published
data and then used it to identify caveats in our current knowledge;
these caveats then were addressed by exploiting relevant high-
throughput datasets.
Our integrative modeling approach enabled us to clarify several
aspects of the regulatory network controlling lymphoid and mye-
loid cell specification. First, although E2a was known to be a
master regulator of lymphoid cell specification [required for both
B- and T-cell specification (38)], the mechanism of its activation
remained unclear, as did the mechanism of its repression in mye-
loid cells. In this respect, our analysis points to Ikaros as a main
activator that is itself activated by Mef2c during lymphoid differ-
entiation and repressed by Cebpa during myeloid differentiation.
In our model, Flt3 is considered a mere marker of multipotent/
lymphoid progenitors. Although the Flt3 pathway has been shown
to be required for lymphoid development and, more particularly, for
the expansion of the CLP population, its impact on cell fate (i.e.,
beyond proliferation and cell survival) remains unclear. Likewise,
ectopic Flt3 signaling has been shown to inactivate C/EBPα through
posttranslational modifications (39), but it is unclear whether this
inactivation occurs in physiological conditions.
The Egr2 and Gfi1 cross-inhibitory circuit has been shown to be
important in the early decision between macrophage and B-cell
fates (21). Our analysis suggests that this circuit becomes irrelevant
after B-cell commitment, enabling high expression of Egr2 in both
pre-B and mature B cells. We therefore proposed that Pax5 can act
as an activator of both factors, allowing their coexpression, al-
though it is possible that other factors are involved also.
Concerning the regulation of Cebpa, our work emphasizes the
absence of known repressors in lymphoid cells. Ebf1 has been
proposed to fulfill this function (40). However, the facts that CLPs
lack myeloid potential and show no Cebpa expression and that a
depletion of IL7R impedes the activation of Ebf1 but still allows
B-cell specification until the pre-B stage (which is devoid of mye-
loid potential) suggest that another factor acting more upstream
represses Cebpa. Mef2c has been shown to counteract myeloid
potential (15), but we could not detect any binding at the Cebpa
locus. We therefore proposed Foxo1 as a candidate repressor.
Thus, according to our model, commitment during normal differ-
entiation of MPs would be controlled mainly by the Cebpa–Foxo1
cross-inhibitory circuit. Hence, Foxo1−/−CLPs could show some
myeloid potential. However, other factors could be involved also.
In particular, the delay in Cebpa re-expression during reprogram-
ming (long after Foxo1 inactivation) suggests an additional mech-
anism, possibly involving epigenetic modifications.
Materials and Methods
ChIP-Seq Meta-Analysis. ChIP-seq data were collected from public databases
(Gene Expression Omnibus), and SRR (sequenced reads run) accession numbers
were gathered in Dataset S2 and were automatically downloaded using the
Aspera Connect browser plug-in. SRA (Sequence Read Archive format) files
were converted in FASTQ using fastq-dump and were mapped onto the mouse
mm10 genome using STAR version 2.4.0f1 (41) (see parameters in SI Materials
and Methods). Duplicated reads were removed using picard (broadinstitute.
github.io/picard/). Bigwig tracks were made using Deeptools bamcoverage (42).
Peak calling was performed using macs2 (43). Gene domains were defined as in
ref. 23, and promoter regions were defined as the TF start site −5 kb/+1 kb,
extended up to the next promoter regions or up to 1 Mb in the absence of other
promoter regions. Peaks to gene domain associations were performed using R.
Gene Network Modeling and Simulations. The logical model of hematopoietic
cell specification was built using GINsim version 2.9 software (44), which is freely
available from ginsim.org. All logical simulations (leading to state transition
graphs and hierarchical transition graphs) and computation of stable states
were performed with GINsim. Stochastic simulations of cell populations were
performed using MaBoSS (31). More detailed information can be found in SI
Materials and Methods. The model can be downloaded from the BioModels
database under accession number 1610240000 and from the logical model
repository on GINsim website (ginsim.org).
Constitutive expression
Pulse
Knock-down
Cebpa
SpI1
Cebpb
E2a
Ebf1
Foxo1
Gfi1
Pax5
Genotype
Phenotype
Model
Experiment
Ref
all 0
B cell or all 0
Mac
Mac
Mac
Mac
Mac
Mac
B cell or Mac
B cell or Mac
B cell or Mac
B cell or Mac
Bcell or mixed state
B cell or Mac
Mac
Mac
Mac
Dead
?
Mac
?
?
Mac (24h pulse)
Mac
Mac
Mac
Mac (24h pulse)
?
?
B-cell (24h pulse)
B cell or mixed state
5
26
26
36
B cell (24h pulse)
36
26
26
26
26
26
26
26
Fig. 7.
Table summarizing the impact of selected perturbations (knockin or
knockout) on B-cell transdifferentiation into macrophages (Mac) upon either
a permanent or a transient induction of C/EBPα. Orange boxes represent
macrophages, blue boxes B cells, gray boxes all 0 stable states or cell death.
Two-color boxes denote alternative outcomes (stable states).
5798
|
www.pnas.org/cgi/doi/10.1073/pnas.1610622114
Collombet et al.
Cell Culture. HAFTL (pre-B) cells and the C/EBPα-ER–containing cell derivative
C10 were grown in Roswell Park Memorial Institute (RPMI) medium with
L-glutamine supplemented with 10% (vol/vol) FBS, 1× penicillin/streptomycin,
and 50 μM β-mercaptoethanol. The RAW 264.7 (ATCC TIB-71) macrophage cell
line was grown in DMEM with L-glutamine supplemented with 10% FBS and
1× penicillin/streptomycin.
Western Blot. Western blots were performed using C10 cells and RAW cells as
previously described (22). More information can be found in SI Materials and
Methods. The following antibodies were used at dilution of 1:1,000: Gfi1 (6C5
ab21061; Abcam), Egr2 (EPR4004 ab108399; Abcam), Egr1 (s-25, sc-101033;
Santa Cruz), and GAPDH (6C5 sc-32233; Santa Cruz).
ChIP-Seq. ChIP-seq experiments were performed as described previously (45).
DNA libraries were prepared using Illumina reagents and instructions and
were sequenced on an Illumina Hi-Seq 2000 system. Data are available on the
Gene Expression Omnibus (GEO) database under accession codes GSE86420
(Ebf1 and Foxo1 ChIP-seq) and GSM1290084 (previously published Cebpa ChIP-
seq in Cebpa-induced B cells).
Ectopic Expression of TFs and Gene-Expression Quantitative PCR. Forced ex-
pression of the B-cell TF Foxo1 in RAW cells was performed using retrovirus.
More information can be found in SI Materials and Methods.
ACKNOWLEDGMENTS. We thank the staff of the computing platform at
the Institut de Biologie de l’Ecole Normale Supérieure for support in hard-
ware and software maintenance; the flow cytometry facility at the Cen-
ter for Genomic Regulation (CRG)/Universitat Pompeu Fabra for help
with cell sorting; the genomics facility of the CRG for sequencing;
and Anna Niarakis, Ralph Stadhouders, and Tian Tian for helpful com-
ments regarding earlier versions of this paper. S.C. is supported by a
scholarship from the French Ministry of Superior Education and Research.
J.L.S.O. was supported by a grant from the Ministry of Economy, Industry,
and Competitiveness (MINECO) (IJCI-2014-21872). B.D.S. was supported by
a long-term fellowship from the European Molecular Biology Organiza-
tion (EMBO) (#ALTF 1143-2015). The T.G. laboratory was supported by
Grant 282510 from the European Union Seventh Framework Program
BLUEPRINT and Fundacio la Marato TV3. This work also was supported
by the Spanish Ministry of Economy and Competitiveness, Centro de
Excelencia Severo Ochoa 2013–2017 and the Centre de Rerserca de
Cataluna (CERCA) Programme, Generalitat de Catalunya.
1. Orkin SH, Zon LI (2008) Hematopoiesis: An evolving paradigm for stem cell biology.
Cell 132(4):631–644.
2. Graf T, Enver T (2009) Forcing cells to change lineages. Nature 462(7273):587–594.
3. Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouse
embryonic and adult fibroblast cultures by defined factors. Cell 126(4):663–676.
4. Laiosa CV, Stadtfeld M, Graf T (2006) Determinants of lymphoid–myeloid lineage
diversification. Annual Rev Immunol 24:705–738.
5. Xie H, Ye M, Feng R, Graf T (2004) Stepwise reprogramming of B cells into macro-
phages. Cell 117(5):663–676.
6. Zhang P, et al. (2004) Enhancement of hematopoietic stem cell repopulating capacity
and self-renewal in the absence of the transcription factor C/EBP alpha. Immunity
21(6):853–863.
7. Bonzanni N, et al. (2013) Hard-wired heterogeneity in blood stem cells revealed using
a dynamic regulatory network model. Bioinformatics 29(13):i80–i88.
8. Abou-Jaoudé W, et al. (2015) Model checking to assess T-helper cell plasticity. Front
Bioeng Biotechnol 2:86.
9. Krumsiek J, Marr C, Schroeder T, Theis FJ (2011) Hierarchical differentiation of mye-
loid progenitors is encoded in the transcription factor network. PLoS One 6(8):e22649.
10. Schütte J, et al. (2016) An experimentally validated network of nine haematopoietic
transcription factors reveals mechanisms of cell state stability. eLife 5:e11469.
11. Naldi A, et al. (2009) Logical modelling of regulatory networks with GINsim 2.3.
Biosystems 97(2):134–139.
12. McKercher SR, et al. (1996) Targeted disruption of the PU.1 gene results in multiple
hematopoietic abnormalities. EMBO J 15(20):5647–5658.
13. Yoshida T, Ng SY-M, Zuniga-Pflucker JC, Georgopoulos K (2006) Early hematopoietic
lineage restrictions directed by Ikaros. Nat Immunol 7(4):382–391.
14. Ikawa T, Kawamoto H, Wright LYT, Murre C (2004) Long-term cultured E2A-deficient
hematopoietic progenitor cells are pluripotent. Immunity 20(3):349–360.
15. Stehling-Sun S, Dade J, Nutt SL, DeKoter RP, Camargo FD (2009) Regulation of lymphoid
versus myeloid fate ‘choice’ by the transcription factor Mef2c. Nat Immunol 10(3):
289–296.
16. Zandi S, et al. (2008) EBF1 is essential for B-lineage priming and establishment of a tran-
scription factor network in common lymphoid progenitors. J Immunol 181(5):3364–3372.
17. Horcher M, Souabni A, Busslinger M (2001) Pax5/BSAP maintains the identity of B cells
in late B lymphopoiesis. Immunity 14(6):779–790.
18. Guo H, Ma O, Speck NA, Friedman AD (2012) Runx1 deletion or dominant inhibition
reduces Cebpa transcription via conserved promoter and distal enhancer sites to favor
monopoiesis over granulopoiesis. Blood 119(19):4408–4418.
19. Krysinska H, et al. (2007) A two-step, PU.1-dependent mechanism for developmentally
regulated chromatin remodeling and transcription of the c-fms gene. Mol Cell Biol 27(3):
878–887.
20. Mossadegh-Keller N, et al. (2013) M-CSF instructs myeloid lineage fate in single
haematopoietic stem cells. Nature 497(7448):239–243.
21. Laslo P, et al. (2006) Multilineage transcriptional priming and determination of al-
ternate hematopoietic cell fates. Cell 126(4):755–766.
22. Spooner CJ, Cheng JX, Pujadas E, Laslo P, Singh H (2009) A recurrent network in-
volving the transcription factors PU.1 and Gfi1 orchestrates innate and adaptive im-
mune cell fates. Immunity 31(4):576–586.
23. McLean CY, et al. (2010) GREAT improves functional interpretation of cis-regulatory
regions. Nat Biotechnol 28(5):495–501.
24. Zarnegar MA, Rothenberg EV (2012) Ikaros represses and activates PU.1 cell-type-
specifically through the multifunctional Sfpi1 URE and a myeloid specific enhancer.
Oncogene 31(43):4647–4654.
25. Di Stefano B, et al. (2016) C/EBPα creates elite cells for iPSC reprogramming by up-
regulating Klf4 and increasing the levels of Lsd1 and Brd4. Nat Cell Biol 18(4):371–381.
26. van Oevelen C, et al. (2015) C/EBPα activates pre-existing and de novo macrophage
enhancers during induced pre-B cell transdifferentiation and myelopoiesis. Stem Cell
Rep 5(2):232–247.
27. Jones LC, et al. (2002) Expression of C/EBPbeta from the C/ebpalpha gene locus is
sufficient for normal hematopoiesis in vivo. Blood 99(6):2032–2036.
28. Bérenguier D, et al. (2013) Dynamical modeling and analysis of large cellular regu-
latory networks. Chaos 23(2):025114.
29. Di Tullio A, et al. (2011) CCAAT/enhancer binding protein alpha (C/EBP(alpha))-
induced transdifferentiation of pre-B cells into macrophages involves no overt ret-
rodifferentiation. Proc Natl Acad Sci USA 108(41):17016–17021.
30. Mansson R, et al. (2012) Positive intergenic feedback circuitry, involving EBF1 and
FOXO1, orchestrates B-cell fate. Proc Natl Acad Sci USA 109(51):21028–21033.
31. Stoll G, Viara E, Barillot E, Calzone L (2012) Continuous time Boolean modeling for
biological signaling: Application of Gillespie algorithm. BMC Syst Biol 6(1):116.
32. Ye M, et al. (2003) Hematopoietic stem cells expressing the myeloid lysozyme gene
retain long-term, multilineage repopulation potential. Immunity 19(5):689–699.
33. Zhang DE, et al. (1997) Absence of granulocyte colony-stimulating factor signaling
and neutrophil development in CCAAT enhancer binding protein alpha-deficient
mice. Proc Natl Acad Sci USA 94(2):569–574.
34. Nutt SL, Heavey B, Rolink AG, Busslinger M (1999) Commitment to the B-lymphoid
lineage depends on the transcription factor Pax5. Nature 401(6753):556–562.
35. Ye M, Ermakova O, Graf T (2005) PU.1 is not strictly required for B cell development
and its absence induces a B-2 to B-1 cell switch. J Exp Med 202(10):1411–1422.
36. Bussmann LHL, et al. (2009) A robust and highly efficient immune cell reprogramming
system. Cell Stem Cell 5(5):554–566.
37. Dunn S-J, Martello G, Yordanov B, Emmott S, Smith AG (2014) Defining an essential
transcription factor program for naïve pluripotency. Science 344(6188):1156–1160.
38. Xu W, et al. (2013) E2A transcription factors limit expression of Gata3 to facilitate T
lymphocyte lineage commitment. Blood 121(9):1534–1542.
39. Radomska HS, et al. (2006) Block of C/EBP α function by phosphorylation in acute
myeloid leukemia with FLT3 activating mutations. J Exp Med 203(2):371–381.
40. Pongubala JMRJ, et al. (2008) Transcription factor EBF restricts alternative lineage op-
tions and promotes B cell fate commitment independently of Pax5. Nat Immunol 9(2):
203–215.
41. Dobin A, et al. (2013) STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 29(1):
15–21.
42. Ramírez F, Dündar F, Diehl S, Grüning BA, Manke T (2014) deepTools: A flexible platform
for exploring deep-sequencing data. Nucleic Acids Res 42(Web Server issue, W1):W187–91.
43. Zhang Y, et al. (2008) Model-based analysis of ChIP-Seq (MACS). Genome Biol 9(9):
R137.
44. Chaouiya C, Naldi A, Thieffry D (2012) Logical modelling of gene regulatory networks
with GINsim. Methods Mol Biol 804:463–479.
45. van Oevelen C, et al. (2008) A role for mammalian Sin3 in permanent gene silencing.
Mol Cell 32(3):359–370.
46. Roessler S, et al. (2007) Distinct promoters mediate the regulation of Ebf1 gene ex-
pression by interleukin-7 and Pax5. Mol Cell Biol 27(2):579–594.
47. Lara-Astiaso D, et al. (2014) Immunogenetics. Chromatin state dynamics during blood
formation. Science 345(6199):943–949.
48. Chen X, et al. (1997) Impaired generation of bone marrow B lymphocytes in mice
deficient in C/EBP β. Blood 90(1):156–164.
49. Didier G, Remy E, Chaouiya C (2011) Mapping multivalued onto Boolean dynamics.
J Theor Biol 270(1):177–184.
Collombet et al.
PNAS
|
June 6, 2017
|
vol. 114
|
no. 23
|
5799
SYSTEMS BIOLOGY
COLLOQUIUM
PAPER
|
28584084
|
Runx1 = ( Spi1 )
Ebf1 = ( Ebf1_gene )
E2A_gene = ( Ikzf1 AND ( ( ( Spi1 ) ) ) )
Egr1 = ( Spi1_2 ) OR ( Spi1 AND ( ( ( NOT Gfi1 ) ) ) ) OR ( Pax5_protein_active )
Cd19 = ( CD19_gene )
IL7r_activated = ( IL7 AND ( ( ( IL7r ) ) ) )
CD19_gene = ( Pax5_protein_active AND ( ( ( NOT Cebpa ) ) ) )
Spi1 = ( Spi1 AND ( ( ( Runx1 ) AND ( ( ( NOT Cebpa AND NOT Csf1r_activated ) ) OR ( ( Gfi1 ) ) OR ( ( NOT Cebpb AND NOT Csf1r_activated ) ) ) ) ) ) OR ( Foxo1 AND ( ( ( Ikzf1 AND Ebf1 ) AND ( ( ( NOT Runx1 OR NOT Spi1 ) ) ) ) ) )
Ikzf1 = ( Pax5_protein_active ) OR ( Mef2c )
Pax5_gene = ( Ebf1 AND ( ( ( NOT Cebpa OR NOT Cebpb ) ) ) )
Cebpa = ( Cebpa ) OR ( Cebpa_gene )
Foxo1_gene = ( E2A_protein_active AND ( ( ( NOT Cebpa OR NOT Cebpb ) ) ) )
Spi1_2 = ( Runx1 AND ( ( ( Csf1r_activated AND Spi1 ) AND ( ( ( Cebpa OR Cebpb ) ) ) ) ) )
Cebpb = ( Spi1_2 AND ( ( ( Cebpa OR Cebpb ) ) ) )
E2A_protein_active = ( E2A AND ( ( ( NOT Id2 ) ) ) )
Ebf1_gene = ( E2A_protein_active AND ( ( ( Ebf1 AND Ets1 AND Pax5_protein_active AND Foxo1 ) AND ( ( ( NOT Cebpa OR NOT Cebpb ) ) ) ) ) )
IL7r = ( Spi1 AND ( ( ( Mef2c ) AND ( ( ( NOT Cebpa OR NOT Cebpb ) ) ) ) ) ) OR ( Ebf1 AND ( ( ( Foxo1 ) AND ( ( ( NOT Cebpa OR NOT Cebpb ) ) ) ) ) )
Mac1_gene = ( Spi1 )
Flt3 = ( Spi1 AND ( ( ( NOT Pax5_protein_active ) AND ( ( ( Ikzf1 ) ) ) ) ) )
Cebpa_gene = ( Spi1 AND ( ( ( Runx1 ) AND ( ( ( NOT Foxo1 ) ) ) ) ) )
Gfi1 = ( Ikzf1 AND ( ( ( NOT Egr1 ) ) ) ) OR ( Cebpa AND ( ( ( NOT Egr1 ) ) ) ) OR ( Pax5_protein_active )
Pax5 = ( Pax5_gene )
Foxo1 = ( E2A )
Id2 = ( Cebpb AND ( ( ( NOT Ebf1 AND NOT Gfi1 ) AND ( ( ( Cebpa AND Spi1 ) ) ) ) ) )
E2A = ( E2A_gene )
Mef2c = ( Spi1 AND ( ( ( NOT Cebpa OR NOT Cebpb ) ) ) )
Csf1r_activated = ( Csf1r AND ( ( ( Csf1 ) ) ) )
Csf1r = ( Spi1 AND ( ( ( NOT Pax5_protein_active ) ) ) )
Mac1 = ( Mac1_gene )
Pax5_protein_active = ( Pax5 AND ( ( ( NOT Id2 ) ) ) )
Ets1 = ( E2A_protein_active )
|
RESEARCH ARTICLE
A model of the onset of the senescence
associated secretory phenotype after DNA
damage induced senescence
Patrick Meyer1,2☯, Pallab Maity1,2☯, Andre Burkovski3,4☯, Julian Schwab3,4,
Christoph Mu¨ssel3, Karmveer Singh1,2, Filipa F. Ferreira1, Linda Krug1,2, Harald J. Maier2,
Meinhard Wlaschek1,2, Thomas Wirth5, Hans A. Kestler2,3☯‡*, Karin Scharffetter-
Kochanek1,2☯‡
1 Department of Dermatology and Allergic Diseases, University of Ulm, Germany, 2 Aging Research Center
(ARC), University of Ulm, Germany, 3 Institute of Medical Systems Biology, University of Ulm, Germany,
4 International Graduate School in Molecular Medicine, University of Ulm, Germany, 5 Institute of
Physiological Chemistry, University of Ulm, Germany
☯These authors contributed equally to this work.
‡ These authors are joint senior authors on this work.
* hans.kestler@uni-ulm.de
Abstract
Cells and tissues are exposed to stress from numerous sources. Senescence is a protective
mechanism that prevents malignant tissue changes and constitutes a fundamental mecha-
nism of aging. It can be accompanied by a senescence associated secretory phenotype
(SASP) that causes chronic inflammation. We present a Boolean network model-based
gene regulatory network of the SASP, incorporating published gene interaction data. The
simulation results describe current biological knowledge. The model predicts different in-sili-
co knockouts that prevent key SASP-mediators, IL-6 and IL-8, from getting activated upon
DNA damage. The NF-κB Essential Modulator (NEMO) was the most promising in-silico
knockout candidate and we were able to show its importance in the inhibition of IL-6 and
IL-8 following DNA-damage in murine dermal fibroblasts in-vitro. We strengthen the specu-
lated regulator function of the NF-κB signaling pathway in the onset and maintenance of the
SASP using in-silico and in-vitro approaches. We were able to mechanistically show, that
DNA damage mediated SASP triggering of IL-6 and IL-8 is mainly relayed through NF-κB,
giving access to possible therapy targets for SASP-accompanied diseases.
Author summary
The senescence associated secretory phenotype is developed by cells undergoing perma-
nent cell cycle arrest. This phenotype is characterized by the secretion of a variety of fac-
tors that facilitate tissue breakdown and inflammation and is therefore theorized to, in
part, be causal for aging and age-related diseases. In recent years the SASP has been impli-
cated in a variety of chronic inflammatory diseases. Due to these advances, it is imperative
to better understand the dynamics of this cellular phenotype and to find ways to disrupt
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
1 / 30
a1111111111
a1111111111
a1111111111
a1111111111
a1111111111
OPEN ACCESS
Citation: Meyer P, Maity P, Burkovski A, Schwab J,
Mu¨ssel C, Singh K, et al. (2017) A model of the
onset of the senescence associated secretory
phenotype after DNA damage induced senescence.
PLoS Comput Biol 13(12): e1005741. https://doi.
org/10.1371/journal.pcbi.1005741
Editor: Paola Vera-Licona, University of
Connecticut Health Center, UNITED STATES
Received: November 28, 2016
Accepted: August 22, 2017
Published: December 4, 2017
Copyright: © 2017 Meyer et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which
permits unrestricted use, distribution, and
reproduction in any medium, provided the original
author and source are credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information
files. This plasmid (pCAG-Cre-T2A-mRuby2) can
be obtained from the authors on request and was
deposited in the Addgene repository (Accession ID
102989).
Funding: KS-K is supported by the German
Research Foundation (DFG, SCHA411/15-2) within
the Clinical Research Group KFO142 “Cellular and
Molecular Mechanisms of Ageing – From
Mechanisms to Clinical Perspectives”, also by the
it. We have developed a Boolean network incorporating the major signaling pathways of
the SASP that allows us to specifically investigate interactions of the pathways and genes
involved. We validated our model by reliably reproducing published data on the SASP.
We utilized our model to uncover components that directly control the detrimental effects
of the senescence associated secretory phenotype that are largely caused by IL-6 and IL-8,
two major factors of the SASP in establishing and spreading senescence as well as causing
local inflammation. In subsequent in-vitro experiments, we were able to verify our
computational results and could suggest NEMO as one potential target for therapy of
SASP-related diseases.
Introduction
Age-related diseases can be held accountable for the major part of morbidity and mortality in an
ageing population. Additionally they cause a large proportion of yearly health costs [1]. Cellular
senescence is one of the most prominent events that is likely to contribute to ageing. It refers to
the irreversible cell cycle arrest that is essential when cells encounter detrimental changes. Once
in permanent arrest, these cells are normally cleared by the immune system before they are able
to do any harm to the organism [2]. However, some of these cells persist and develop a secretory
phenotype releasing a variety of factors among which pro-inflammatory cytokines, chemokines
and extracellular matrix degrading proteases are included. Together these shape the senescent-
associated secretory phenotype or SASP [3–5].
While the SASP can cause chronic inflammation in tissue, it can also reinforce senescence
in autocrine and paracrine manner [6, 7]. This feature of the SASP not only keeps senescent
cells in their growth arrested states but it promotes senescence spreading to healthy bystander
cells. Therefore, the SASP contributes to the accumulation of senescent cells during ageing,
but also supports the emergence of age-related chronic diseases and tissue dysfunctions by
elevating inflammatory processes [6, 8]. Major soluble factors that facilitate this bystander-
infection of healthy cells are IL-6 and IL-8. Both have been shown to be important in the main-
tenance and spreading of oncogene- and DNA-damage-induced senescence [3]. Also, both
have been shown to be highly overexpressed by senescent cells and are known to locally and
systemically play important roles in the regulations of a variety of processes in the aging body
[3, 4, 9]. IL-6, in fact, most likely contributes to organ dysfunction during aging thus promot-
ing frailty [8].
To allow for a deeper understanding of the SASP and the dynamics of its complex interac-
tions a computational model of the Regulatory Network (RN) [10] and subsequent simulations
can be insightful. RNs can be described by different mathematical models such as differential
equations, Bayesian networks, and Boolean networks among others [11]. The Boolean network
model [12, 13], as opposed to other model approaches, can be based on qualitative knowledge
only. In gene-gene interaction, for example, the expression of a gene is regulated by transcrip-
tion factors binding to its regulatory regions. The activation of a gene follows a switch-like
behavior depending on the concentration of its transcription factors. This behavior allows
common approximation of the possible states of a gene to be active or inactive [14, 15]. Ulti-
mately, this can be encoded as Boolean logical values: true (“1”) or false (“0”). The interactions
between genes, e.g. whether a factor acts as an activator, repressor or both can be described by
functions. These Boolean functions are the basis to simulate dynamic behavior, i.e. changes
over time. As every regulatory factor has two possible states (active or inactive) in a Boolean
network model, 2x possible state combinations (i.e. gene activation patterns) exist for x genes.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
2 / 30
Graduate Training Centre GRK 1789 “Cellular and
Molecular Mechanisms in Ageing (CEMMA)”, and
collaborative Project FKZ0315894A SyStaR -
Molecular Systems Biology of Impaired Stem Cell
Function and Regeneration during Aging, and
Collaborative Research Centre CRC1149 Danger
Response, Disturbance Factors and Regenerative
Potential after Acute Trauma and the Fo¨rderlinie
Perspektivfo¨rderung “Zellula¨re Entscheidungs- und
Signalwege bei der Alterung” of the Ministerium fu¨r
Wissenschaft, Forschung und Kunst Baden-
Wu¨rttemberg, Germany. HAK is supported by the
European Community’s Seventh Framework
Programme (FP7/2007–2013) under grant
agreement n602783, the DFG (SFB 1074 project
Z1), and the German Federal Ministry of Education
and Research (BMBF, Gerontosys II,
Forschungskern SyStaR, project ID 0315894A and
e:Med, SYMBOL-HF, ID 01ZX1407A)
Competing interests: The authors have declared
that no competing interests exist.
For any activation pattern, iterative updates of genes in the network through consecutive appli-
cation of the Boolean rules eventually lead to sequences of gene activation patterns that are
time-invariant, called attractors. These attractors can correspond to observed expression pro-
files of biological phenotypes or can be used to create hypotheses to further evaluate in wet-lab
experiments [16, 17]. Different update strategies for the Boolean functions exist. Using a syn-
chronous update strategy means applying all Boolean functions simultaneously, also assuming
that regulatory factors interact independently of one another and that their interaction has a
similar time scale resolution. Relaxing these assumptions leads to the concept of asynchronous
updates where each Boolean function of is updated separately one at a time in any order. This
allows a more direct modelling of different time scales. The asynchronous update strategy also
usually generates trajectories that are different from those of synchronous Boolean networks.
The state transition graph of an asynchronous Boolean network becomes a Markov chain
which requires the additional definition of transition probabilities in each node of the state
graph. Interestingly, point attractors (those with one state) in asynchronous Boolean networks
are the same as those in synchronous Boolean networks. However, these networks can also
show loose/complex attractors [18] which are part of active research [19, 20]. Another exten-
sion of Boolean networks are probabilistic Boolean networks, which may define more than
one Boolean function for regulatory factors where each function has a specific probability to
be chosen for update. Although this concept may closer represent a biological system, it again
requires parameter estimation for the probabilities. However, estimation of the probabilities
naturally demands large amounts of interaction specific data which is, for larger networks, nei-
ther economically, nor experimentally viable. In our case, we decided to focus on synchronous
Boolean networks, partly due to their proven usability, and their ability to reveal key dynamical
patterns of the modelled system. However, to strengthen our models’ hypothesis, we addition-
ally performed in-silico experiments with an asynchronous update scheme (S1 Text).
Synchronous Boolean networks have been used to model the oncogenic pathways in neuro-
blastoma [21], the hrp regulon of Pseudomonas syringae [22], the blood development from
mesoderm to blood [23], the determination of the first or second heart field identity [24] as
well as for the modeling of the Wnt pathway [25]. The qualitative knowledge base that is neces-
sary to reconstruct [26] a Boolean network model consists mostly of reports on specific inter-
actions that describe local regulation of genes or proteins. Boolean network models utilize this
knowledge about local regulations to reconstruct a first global mechanistic model of SASP. In
summary, such a model allows to generate hypotheses about regulatory influences on different
local interactions. These interactions, in turn, can be tested in wet-lab in order to validate the
generated hypothesis and assess the accuracy of the proposed model.
Here, we present a regulatory Boolean network of the development and maintenance of
senescence and the SASP incorporating published gene interaction data of SASP-associated
signaling pathways like IL-1, IL-6, p53 and NF-κB. We simulated the model and retrieved
steady states of pathway interactions between p53/p16INK4A steered senescence, IL-1/IL-6
driven inflammatory activity and the emergence and retention of the SASP through NF-κB
and its targets. This Boolean network enables the highlighting of key players in these processes.
Simulations of knock-out experiments within this model go in line with previously published
data. The subsequent validation of generated in-silico results in-vitro was done in murine der-
mal fibroblasts (MDF) isolated from a murine NF-κB Essential Modulator (NEMO)-knockout
system in which DNA damage was introduced. The NEMO knockout inhibits IL-6 and IL-8
homologue mRNA expression and protein secretion in MDFs after DNA damage in-vitro,
possibly enabling at least a lowering of the contagiousness for neighboring cells and the pro-
tumorigenic potential of the SASP. The model presented in this article allows a mechanistic
view on interaction between the proinflammatory and DNA-damage signaling pathways and
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
3 / 30
thereby helps to gain insights into the dynamics of the SASP. Furthermore, it enables to gener-
ate extensive hypotheses about possible knockout targets that can be experimentally tested and
verified in-vitro. To the best of our knowledge, this report is the first one that combined in-sil-
ico simulation of the SASP with its laboratory based experimental validation.
Results
The network model exhibits stable states for cell cycle progression and
senescence
The reconstruction of a Boolean network model for SASP requires screening for many candi-
date interactions in published literature and data. Although the model, after reconstruction,
may be reduced in the number of components [20, 27, 28], it would potentially hide some of
the interaction targets and regulatory factors with regard to the signaling cascade. The regula-
tory factors defined in this model are beneficial if one wants to extend the model and include
additional related signaling pathways. The subsequent model must accurately correspond to
the current understanding of the process at hand, i.e., able to predict well-known phenotypes
of SASP. Biological phenotypes represent a long-term behavior of a biological system based on
interaction of regulatory factors. In the same sense, attractors are the long-term behavior of a
Boolean network model based on the Boolean rules of modelled regulatory factors. Hence,
there is a natural correspondence between biological phenotypes and attractors in the Boolean
network. In the following, we use figures that depict the signaling cascade towards an attractor
as well as the attractor itself. The interpretation of these attractors in the context of SASP fur-
ther allows generation of hypotheses that can be tested in a biological system.
The information for the reconstruction of these networks was collected from published
data. An overview of the genes incorporated in this model and their interaction can be found
in Fig 1. The corresponding Boolean rules are listed in Table 1. The network depicts processes
following a cell cycle arrest inducing action, such as DNA damage and other cellular stresses.
Here, we analyze SASP under strong DNA damage and do not distinguish between different
levels of DNA damage.
We first analyzed if our model can render steady states for cell cycle progression when there
is no stress signal input. Our data show a normal cell cycle progression with active CDK2 and
CDK4, as well as phosphorylated Rb and hence an active E2F. No other signaling pathways
that are implemented in this model were activated which can be seen as normal cell cycle pro-
gression (Fig 2).
Upon the outside signal DNA damage, we observe first the activation of the DNA damage
response with a subsequent activation of p53 and p16INK4A signaling, leading to a stop in cell
cycle progression and at a later time point to permanent cell cycle arrest. Simultaneously NF-κB
signaling gets activated by the DNA damage response through NEMO, giving rise to beneficial
but also detrimental effects of NF-κB like the senescence associated secretory phenotype (Fig 3).
After entering p53/p21 and p16INK4A mediated permanent cell cycle arrest upon DNA
damage, the activation of NF-κB leads to an increase of IL-1, IL-6 as well as IL-8 expression
among others [29–33]. Our model shows the direct activation of these cytokines and chemo-
kines by NF-κB after its activation through the DNA damage response and NEMO (Fig 3).
The Boolean network describes published knock-out and
overexpression phenotypes
The NF-κB pathway has been studied extensively and there are knockout mice available for all
proteins of the pathway, however some of them are embryonically lethal due to the importance
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
4 / 30
of NF-κB signaling in regulating development and apoptosis. We therefore focused on pub-
lished in-vitro knockout and overexpression phenotypes. IL-6 and IL-8 are extremely impor-
tant in maintaining and spreading the SASP in an autocrine as well as paracrine fashion.
Hence, we followed the question what knockouts and/or overexpressions the Boolean network
model suggests to inhibit the expression of IL-6 and IL-8 under the assumption of existing
DNA damage. These simulations are included in S1 Text.
Fig 1. Boolean network for gene regulation during cell cycle progression and the onset of cell cycle arrest after DNA damage. The overview
shows the network wiring of the known gene regulations during DNA damage with a focus on the DNA damage repair/cell cycle arrest signaling. Cell
cycle arrested cells over time show a tendency to develop a secretory phenotype that causes them to secrete high amounts of proinflammatory factors
that can negatively influence neighboring cells. Major signaling pathways of these factors are included in this overview and in the Boolean network.
Arrows indicate gene activation and inhibition is depicted as bar head. However, the interaction may be more complex and the corresponding Boolean
rules are given in Table 1.
https://doi.org/10.1371/journal.pcbi.1005741.g001
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
5 / 30
Table 1. Boolean network for gene regulation during cell cycle progression and the onset of cell cycle arrest after DNA damage. Boolean Rules
using operators “&” (logical and), “|” (logical or) and “¬” (logical not).
DNA Damage/Senescence signaling
Regulatory Factor at
time t+1
Boolean rule update given regulatory
factor state at time t
DNA Damage, Defective Telomeres, etc.
DNAD
DNAD
This rule serves as an input signal to any kind of severe DNA damage.
Oncogene induced senescence
Oncogene IL8 | IL6
Active IL-6 or IL-8 signaling characterize the activation of Oncogene. Moreover, IL-6 and IL-
8 also required for oncogene induced senescence [3].
Hypoxia
Hypoxia
Exogenous factor describing Hypoxia.
In presence of DNA damage, a cell activates regulatory factors ATR and ATM, which subsequently activate checkpoints CHK1 and
CHK2.
ATM
DNAD
ATM is active in presence of DNA damage [57–59].
CHK2
ATM
ATM subsequently activates CHK2 [60].
ATR
DNAD
ATR is active in presence of DNA damage [57, 59].
CHK1
ATR
ATR subsequently activates CHK1 [61].
p53
(CHK2 | CHK1 | ATM) & (¬MDM2)
p53 can be activated by any of CHK1 [62], CHK2 [62, 63] or ATM [62, 64]. However, MDM2
is a strong inhibitor of p53 [62, 65].
HIF1
Hypoxia & (¬p53)
HIF1, which is active during Hypoxia [66], is inhibited by p53 [67].
p21
p53 | HIF1
p21 is activated by p53 [68] as well as by HIF1 [69].
CDK2
E2F & (¬p21)
CDK2 requires activation of E2F. p21 inhibits the CDK2 complex [68].
RB
¬(pRB | CDK4 | CDK2)
RB, which is active in its hypophosphorylated state (RB) is hyperphosphorylated and
inactivated (pRB) by CDK4 and CDK2 [70–72].
pRB
(CDK4 | CDK2)
RB is phosphorylated (pRB) in presence of any cyclin dependent kinases CDK4 and CDK2
[70–72].
E2F
(pRB | E2F) & ¬RB
E2F is positively autoregulated and active in presence of hyperphosphorylated RB (pRB).
Active RB, however, inhibts E2F [38].
MDM2
p53 & ¬ATM
p53 activates MDM2 [65, 73, 74], while ATM inhibits MDM2 [64].
p16INK4 Oncogene | DNAD
Activation of p16INK4 depends on either DNA damage or Oncogene or both [75].
CDK4
¬(p16INK4 | p21)
CDK4 is inhibited by p16INK4 [75] and p21 [68].
NEMO
DNAD
NEMO is activated by DNA damage [76, 77].
IKK
NEMO | NIK | Akt
IKK can be activated by any of NEMO [78], NIK [79] or Akt [80].
IkB
(NFkB |IkB) & ¬(IKK & NEMO)
IkB is activated NFkB complex or IkB itself [81]. IKK [82] and NEMO [83] together are
required to inhibit IkB.
NFkB
IKK & ¬IkB
NFkB is activated by IKK, while inhibited by IkB [82, 83].
IL-1 signaling
IL1
NFkB
IL1 is activated by NFkB [29, 30].
IL1R
IL1
IL1 binds to and activates IL1 receptor (IL1R) [84].
MyD88 IL1R
MyD88 is an adaptor molecule in IL1-IL1R pathway and bridging IL1R to the IRAK complex
IL1R [84].
IRAK
IL1R | MyD88 | IRAK
IRAK is autoactivated [85, 86] and also is activated by IL1R [84, 86] and MyD88 [85, 87].
TRAF6
IRAK
TRAF6 is activated by IRAK [85].
TAB
(TRAF6 | IRAK)
TAB is activated by any of TRAF6 [88, 89] or IRAK [89].
TAK1
(TRAF6 | TAB)
TAK1 is activated by any of TRAF6 [88, 89] or TAB [90].
MEKK
TRAF6
MEKK is activated by TRAF6 [89].
MKK
(TAK1 | MEKK)
MKK is activated by any of TRAK1 [91, 92] or MEKK [93].
JNK
MKK & ¬MKP1
JNK is activated by MKK [94, 95] while is inhibited by MKP1 [96].
p38
MKK & ¬MKP1
p38 is activated by MKK [97] while inhibited by MKP1 [98].
cJun
(p38 | JNK | ERK1_2 | CEBPbeta) &
cFos
cFos is required for the action of cJun and can be activated by any one of p38 [99, 100],
JNK [101], ERK1_2 [102] or CEBPbeta [103].
(Continued)
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
6 / 30
RelA binds with p50 to form a transcriptionally active heterodimer (called NFkB in this
model). In its inactive state, it is bound with the inhibitor of kappa B (IκB) and resides in the
cytoplasm. Upon NF-κB activation, the inhibitor is phosphorylated by the inhibitor of kappa B
kinases (IKK) and degraded which releases the RelA/p50 heterodimer to translocate to the
nucleus and regulate the transcription of target genes. To investigate the role of RelA on the
expression of IL-8, we set NFkB = 0, simulating the ablation of the transcriptionally active het-
erodimer (Fig 4). The predictions of the model simulations are consistent with knock-out
experiments where the absence of RelA caused a significant reduction in IL-8 production in
human fibroblast (IMR-90) [7].
We also simulated the overexpression of IκB by constantly activating IκB (IkB = 1) and
could show an effect comparable to the knock-out of RelA (Fig 5). In our model the overex-
pression of IκB leads to the inhibition of IL-8 and IL-6 expression which is in line with a previ-
ously published report, where the overexpression of a non-degradable IκBα completely
abolishes IL-8 production, among other soluble factors, in human epithelial and cancer cell
lines [34].
Another promising knockout described by our network is inhibitor of nuclear factor
kappa-B kinase subunit gamma also known as NEMO, which is able to prevent IL-6 and IL-8
expression after DNA damage activated the DNA damage repair apparatus and cell cycle pro-
gression has been stopped in-silico (Fig 6). In studies with murine NEMO knockout models it
has already been shown that murine embryonic fibroblasts (MEFs) isolated from these mice
show reduced NF-κB activity and IL-6 secretion upon stimulation with typical NF-κB activa-
tors like IL-1 and TNF [35].
Table 1. (Continued)
DNA Damage/Senescence signaling
cFos
p38 | JNK | Elk1 | CEBPbeta | STAT3
cFos can be activated by any one of p38 [104], JNK [104], Elk1 [103, 105, 106], CEBPbeta
[103] or STAT3 [107].
AP1
cJun & cFos
AP1 complex consists of both cJun and cFos [104, 108].
MPK1
AP1
AP1 activates MPK1 [96, 109, 110].
IL8
NFkB | AP1 | CEBPbeta
IL8 is activated by anyone of NFkB [31, 111, 112], AP1 [31] or CEBPbeta [3] signals.
NIK
TAK1
NIK is activated by TAK1 [91, 92].
IL-6 signaling
IL6
(NFkB | ERK1_2 | CEBPbeta)
IL6 is activated by anyone of NFkB [32, 33], ERK1_2 [113, 114] or CEBPbeta [3, 115]
signals.
IL6R
IL6
IL6 binds to and activates IL6 receptor (IL6R) [88, 116].
GP130
IL6
GP130 is activated by IL6 [117, 118].
PI3K
JAK
PI3K is activated by JAK [119].
JAK
IL6R & ¬SOCS3
Active IL6 receptor (IL6R) activates JAK [117], while JAK is inhibited by SOCS3 [120].
Akt
PI3K
Akt is activated by PI3K [121, 122].
mTOR
Akt
mTOR is activated by Akt [123].
SOCS3
STAT3
SOCS3 is activated by STAT3 [124].
GP130, MEK1_2, and ERK1_2 together depend all on the activation of IsL6 to form a cyclic signaling cascade
MEK1_2 GP130 & IL6
MEK1_2 is activated by GP130 [116, 125] as well as IL6 [116].
ERK1_2
MEK1_2 & IL6
ERK1_2 is activated by MEK1_2 [126] and IL6 [127].
Elk1
ERK1_2
Elk1 is activated by ERK1_2 [128].
CEBPbeta
Elk1
CEBPbeta is activated by Elk1 [103].
STAT3 JAK | (cFos & cJun) | mTOR
STAT3 is activated by JAK [119] or mTOR [129]. Alternatively is can be activated in
presence of both cFos and cJun [130].
https://doi.org/10.1371/journal.pcbi.1005741.t001
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
7 / 30
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
8 / 30
NEMO is essential for DNA damage triggered NF-κB activation
Apart from being important for the assembly of the IKK-complex, NEMO also acts as a shuttle
relaying the ATM-mediated DNA damage apparatus to cellular response mechanisms. Upon
DNA damage ATM can bind NEMO and trigger its translocation from the nucleus to the cyto-
plasm where it activates NF-κB signaling [36]. This in turn will help cells avoid clearance
through apoptosis, increasing the number of long-term senescent cells in tissues and organs of
the organism and might also increase and sustain the inflammatory potential of the SASP.
In order to evaluate proposed knockouts NEMO was depleted from murine dermal fibro-
blasts (MDFs) using a NEMO-floxed mouse line. These MDFs were isolated from murine skin
and subsequently transfected with a Cre-recombinase coding plasmid including a fluorescence
reporter construct (Fig 7). To purify NEMO knockout MDFs, these cells were FACS sorted
two days post-transfection (S1A Fig). Successful NEMO knockout was assessed by PCR (S1B
Fig) and western blot (S1C Fig). To study the effect of DNA damage, overnight-starved MDFs
were treated with 25 μM etoposide, an established DNA damage and senescence inducer, for 3
h followed by a 24 h incubation period [37]. Afterwards cell media supernatant was taken and
total RNA was isolated. We first measured p21 mRNA expression as an indicator for DNA
damage and cell cycle arrest. Without a significant reduction of cell viability (Fig 8A), p21
mRNA expression was upregulated more than twofold in etoposide treated compared to
untreated MDFs (Fig 8B). NEMO is of high importance for DNA damage mediated nuclear
translocation of the NF-κB signaling molecule p65. As shown by immunofluorescence staining
of untreated NEMO wildtype MDFs compared to etoposide treated wildtype and knockout
MDFs, the translocation of p65 into the nucleus upon DNA damage is significantly increased
in wildtype whereas it is brought down to the level of untreated wildtype MDFs when NEMO
is knocked out (Fig 8C).
NEMO mediates DNA damage induced expression and secretion of IL-6
and IL-8
As we have observed the effect of a NEMO knockout on the nuclear translocation of p65 and
thereby activation of NF-κB, we further explored the possible suppressive effect on IL-6 and
IL-8 activation. To achieve this we isolated total RNA and analyzed the mRNA expression of
IL-6 and the murine homologues of IL-8 CXCL1 (KC), CXCL2 (MIP-2) and CXCL5 (LIX).
Upon DNA damage, we observed a significant reduction in IL-6 mRNA expression with a
strong downregulation in untreated knockout compared to untreated wildtype. An even stron-
ger downregulation in etoposide treated NEMO knockout compared to wildtype MDFs was
detected. Taken together a NEMO knockout could reduce DNA-damage mediated IL-6
mRNA expression by almost tenfold (Fig 9A). Next, we measured the secretion of IL-6. While
there is nearly no secretion of IL-6 in untreated wildtype as well as knockout MDFs, a strong
increase in IL-6 secretion occurred in etoposide treated wildtype MDFs, whereas the NEMO
knockout MDFs only shows a small increase in secretion with a more than hundredfold reduc-
tion when compared with etoposide treated wildtype cells (Fig 9B). We additionally analyzed
the mRNA expression of three murine IL-8 homologues to assess the impact of a NEMO
knockout on DNA damage mediated IL-8 expression. We found that all three chosen
Fig 2. Naturally occurring network states. Without DNA damage the resulting network state is expected to
show normal cell cycle progression. As shown here this includes the activation of CDK2 (t = 5) and CDK4
(t = 2) with a subsequent phosphorylation of RB (t = 3) leading to a release of E2F (t = 4) which will release the
cell into cell cycle progression. The temporal sequence is shown as t = n. Active genes are shown as green,
inactive genes as dark purple.
https://doi.org/10.1371/journal.pcbi.1005741.g002
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
9 / 30
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
10 / 30
homologues were significantly downregulated in NEMO knockout MDFs compared to wild-
type MDFs after DNA damage. The total expression of IL-8 homologues mRNA in NEMO
knockout MDFs was reduced by at least fivefold when compared to treated wildtype MDFs
(Fig 9C). There is detectable secretion of IL-8 homologues in untreated wildtype and NEMO
knockout MDFs, however the secretion strongly rose upon etoposide treated in wildtype cells
whereas there is no detectable increase in the NEMO knockout MDFs. This effect was similarly
found for the studied IL-8 homologues KC and MIP-2 (Fig 9D). However, we did not find any
significant alteration in the expression of two housekeeping genes, such as beta-actin and 18s
rRNA in the NEMO knockout MDFs, compared with NEMO wildtype (S2A Fig). In addition,
we also did not observe any significant alteration in the expression of a wide array of genes
that were predicted by Boolean network not to be changed after NEMO knockout (S2B Fig).
These data show the importance of NEMO and NF-κB signaling for the activation of IL-6 and
IL-8 in the case of DNA damage. In early stages DNA damaged and cell cycle arrested MDFs
most likely activate secretory SASP signaling through NF-κB rather than other stress
pathways.
Discussion
In the model of DNA damage and proinflammatory signaling presented here we collected and
combined previously published knowledge on major regulators of the SASP. Using this model,
we identified attractors fitting cell cycle progression and cell cycle arrest as they physiologically
occur. This suggests reliability of this model in terms of reproducibility of current biological
knowledge. The network model allows us to time- and cost-effectively generate hypotheses
and predict gene knockouts that may influence the outcome of the SASP in-vitro.
In the process of modeling, we first created individual models of DNA damage and proin-
flammatory signaling. In a next step, we fused these two sub-networks to the model presented
here. In S1 Text, we analyzed the impact of integrating both pathways in one Boolean network
model. Our results indicate that there is not only an effect of DNA damage in the proinflam-
matory signaling but also vice versa. On one hand, we deduce a stabilization of the DNA dam-
age response network as the integration of both sub-networks leads to a reduction of possible
attractors (87 to 19). On the other hand, the inner dynamics of each sub-network stay intact,
showing biologically reproducible signaling cascades (e.g. Fig 4).
In the simulation without DNA damage, only activation of cell cycle regulation genes that
facilitate cell cycle progression were observed [38]. In contrast, when we entered DNA damage
into the network, we detected early activation of the DNA damage response (DDR) followed
by a p53/p21 mediated cell cycle arrest and at a later time point the activation of proinflamma-
tory signaling through NF-κB [39, 40]. We utilized the Boolean network to simulate knockout
and overexpression states that have the power to inhibit both IL-6 and IL-8 activation, such as
knockouts of ATM and RelA or the overexpression of IκBα, that have previously been pub-
lished to decrease IL-8 or IL-6 expression and secretion in-vitro [7, 9, 34]. One of the most
prominent knockout suggestions obtained was that of NEMO, which acts as an essential mod-
ulator of NF-κB signaling and is a major link between DDR and NF-κB signaling [41]. There-
fore, it is a suitable target to prevent NF-κB activation, while maintaining the repair potential
of the DDR. Taken together these in-silico data suggest NF-κB to be one of the major SASP
Fig 3. Naturally occurring network states upon DNA damage. Upon DNA damage the first response of the cell is
the activation of ATM/ATR mediated DNA damage repair (t = 2) with a subsequent activation of p53- and p16-mediated
cell cycle arrest (t = 3). The DNA damage signal is relayed by the DNA damage response through NEMO (t = 3) that in
turn activates NF-κB signaling (t = 4) which will ultimately lead to the activation of IL-1, IL-6 and IL-8 signaling (t = 7). The
temporal sequence is shown as t = n. Active genes are shown as green, inactive genes as dark purple.
https://doi.org/10.1371/journal.pcbi.1005741.g003
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
11 / 30
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
12 / 30
activators in response to DNA damage activating all three mediators of proinflammatory sig-
naling depicted in this network.
For the sake of manageability, the model presented here was limited to a core set of path-
ways involved in senescence and the SASP. Of course, the value of the results could still be
enriched by adding even more components and additional pathways, such as a more detail
view on CEBP-signaling, growth factor signaling and the expansion of cell cycle related signal-
ing. This would enable to simulate an even deeper level of signaling involved in the SASP.
Another factor that was not viewed in this work is the influence of the intensity levels and tim-
ing of expression and stimuli on the outcome of the SASP. Physiologically occurring DNA
damage, for example, is not an all or nothing event but rather comes in different levels and
lengths of damage that can trigger a multitude of different reactions in the cell. In future
works, it would be interesting to add these into the model. Such extension would allow simula-
tions of the exact amount and timing of damage needed to trigger full-blown SASP rather than
senescence. Furthermore, it would possibly reveal at which point the cell decides that it is ben-
eficial to trigger SASP signaling in order to warn the system of the damage and initiate clear-
ance as opposed to trying to repair itself.
IL-6 and IL-8 reinforce senescence in an autocrine and paracrine way, concomitantly pre-
venting senescent cells from exiting cell cycle arrest and forcing neighboring cells into senes-
cence themselves [3, 42]. Persistent DDR activity, that is also known to induce IL-6 and IL-8
secretion [9], could be shown in various premalignant and malignant lesions in-vivo, and is
hypothesized to be one the main causes of aging [9, 43, 44]. Due to this ability to promote inva-
siveness of cancer cells and the spreading of senescence to neighboring cells IL-6 and IL-8 are
of special interest [3, 45]. While it is probably not detrimental to transiently activate the respec-
tive signaling pathways, the long-term persistence of unrepairable DNA damage leads to a
lasting activation of NF-κB through the DDR mechanisms and thereby to a prolonged stimula-
tion of IL-6 and IL-8. Ultimately, this initiates and perpetuates a vicious cycle from which cells
cannot escape and causes the development of the SASP.
To explore and validate previously generated in-silico results in-vitro, we isolated murine
dermal fibroblasts from NEMO-floxed mice and transfected these with a Cre-recombinase
plasmid to deplete NEMO. Contrary to NEMO knockout MDFs we observed RelA enrichment
in the nucleus in DNA damaged wildtype cells. This suggests that mainly NEMO is responsible
for the forwarding of DNA damage signals from the DDR to NF-κB signaling.
We were particularly interested in achieving inhibition of IL-6 and IL-8 expression and
secretion in-silico and in-vitro. As we could show in our in-vitro results, DNA damaged NEMO
knockout cells did not reveal any induction of IL-6 or IL-8 homologue mRNA expression, sug-
gesting that DNA damage-triggered IL-6 and IL-8 expression is mainly conferred by NF-κB sig-
naling. This was confirmed on protein level, showing a strong decrease in secretion of both IL-6
and IL-8 homologues in NEMO knockout MDFs. In conclusion, abolishing NEMO is sufficient
to not only block the signaling from DDR to NF-κB but also to decrease expression and secre-
tion of two of the most prominent and established SASP mediators IL-6 and IL-8.
The question arises why damaged senescent cells have to start expressing and secreting fac-
tors that are detrimental to themselves, surrounding cells and tissues. The secretion of many
SASP factors can be explained firstly by the attempt to clear senescent cells from tissue by cells
Fig 4. Knockouts that cause in-silico IL-6 and IL-8 inhibition for NFkB knockout. Network states present
the gene activity of all genes in the model. Green boxes indicate gene activation while red boxes show gene
inactivation. A knock-down or overexpression is simulated by setting a gene to 0 or 1, respectively. This
simulation shows the time course of expected states after DNA damage with NF-κB switched off (NFkB = 0)
which leads to an inhibition of proinflammatory signaling.
https://doi.org/10.1371/journal.pcbi.1005741.g004
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
13 / 30
Fig 5. Knockouts that cause in-silico IL-6 and IL-8 inhibition for IkB overexpression. This simulation
shows an overexpression of IκB (IkB = 1) showing a similar outcome as in Fig 4.
https://doi.org/10.1371/journal.pcbi.1005741.g005
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
14 / 30
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
15 / 30
of the innate immune system and secondly as a warning to the microenvironment that there is
a danger in the near vicinity. Senescent cells secrete different factors that attract phagocytic
immune cells and induce proteolytic enzymes to facilitate their migration through the extracel-
lular matrix [46]. As long as damaged cells can be cleared in early phases the SASP is probably
beneficial for the organism, however once the immune system cannot keep up with the emer-
gence of damaged cells, detrimental effects accumulate and tissue takes damage [2, 47]. In this
phase, it would be beneficial to have the possibility to counteract the SASP and give the
immune system time to catch up.
In summary, we could illustrate that in-silico identification of genes with mechanistic con-
tribution in the regulation of the SASP, confirmed under experimental conditions in-vitro, is a
highly suitable approach and holds substantial promise to identifying therapeutic targets to
delay or even prevent the detrimental SASP effects on tissue homeostasis and overall ageing.
Using our Boolean model, we were able to reproduce published data in-silico and generate var-
ious knockout proposals to shut down two of the most detrimental effectors of the SASP. This
is of major clinical relevance in terms of tissue aging. In fact, SASP factors like IL-6 and IL-8
have been correlated with inflammaging not only driving the aging process itself, but also
Fig 6. Knockouts that cause in-silico IL-6 and IL-8 inhibition for NEMO knockout. NEMO is switched off
(NEMO = 0) preventing NF-κB signaling from being activated. The outcome is similar to the two previously
described simulations in Figs 4 and 5.
https://doi.org/10.1371/journal.pcbi.1005741.g006
Fig 7. Schematic overview of the experimental workflow. Murine dermal fibroblasts (MDFs) are isolated
from NEMO-floxed mice. After short expansion in cell culture these MDFs are transfected with pCAG-Cre-
T2A-mRuby2 or pCAG-mRuby2, respectively. Because of mRuby2 expression, successfully transfected cells
can be sorted by FACS. Cells transfected with pCAG-Cre-T2A-mRuby2 are knocked out for NEMO while
pCAG-mRuby2 transfected cells are used as wildtype controls. After transfection cells are treated with 25 μM
etoposide for 3 h to induce DNA damage. 24 h after treatment cell culture media is taken for ELISA
measurement of secretion and cells are harvested for RNA isolation and subsequent RT-qPCR analysis.
https://doi.org/10.1371/journal.pcbi.1005741.g007
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
16 / 30
promoting aging associated morbidity, frailty and mortality [48]. We additionally were able to
validate and prove one of the most prominent knockout suggestions in-vitro, keeping in mind
that there might always be detrimental off-target effects when altering a major signaling path-
way like NF-κB. However, targeting NEMO and its interaction partners, as already shown in
Fig 8. NEMO knockout murine dermal fibroblasts show a decreased nuclear translocation of p65. a. MTT assay
determined optimal experimental conditions. 80% viable cells was set as threshold. After overnight serum starvation MDFs were
treated with etoposide for 3 h followed by a 24 h incubation period. MTT assay was started afterwards to determine the viability
of cells. Values are presented as mean ± SEM in percent. (n = 3) b. In order to evaluate DNA damage response and cell cycle
arrest mRNA expression of p21 was analysed by RT-qPCR in MDFs treated with 25μM etoposide for 3 h followed by a 24 h
incubation time (n = 5). Values are presented as mean ± SEM of fold change. Comparison was made with two-tailed t-test; P-
value indicated the significance of difference. c. Representative immunostaining of γH2Ax (green) and p65 (red) in wildtype
(NEMO WT) and NEMO knockout (NEMO k/o) MDFs treated with 25μM etoposide for 3 h with a following incubation period of
24 h. Scale bars, 50μM. The graph shows the percentage of p65 in the cytoplasm (black bars) compared to the nucleus (grey
bars) as percentage of red pixels. Values are mean ± SEM in percent. Comparison was made with two-tailed t-test (n = 10); line
and P-value.
https://doi.org/10.1371/journal.pcbi.1005741.g008
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
17 / 30
studies of inflammatory arthritis and diffuse large B-cell lymphoma, may hold promise for the
development of new therapies for age-related pathologies in which senescence and the SASP
play a role [49, 50].
Fig 9. DNA damaged NEMO knockout MDFs show a decrease in IL-6 and IL-8 mRNA expression and
protein secretion. a. To assess the influence of the NEMO knockout on DNA damage mediated activation of
SASP signaling IL-6 mRNA expression was measured by RT-qPCR in untreated and etoposide-treated MDFs
(n = 5). Cells with wildtype NEMO (black bars) or NEMO knockout (grey bars) were used. Values were presented
as mean ± SEM of fold change. Comparison was made with the two-tailed t-test. b. IL-6 secretion was measured
by ELISA in conditioned media of untreated and etoposide-treated MDFs (n = 5). Cells with wildtype NEMO (black
bars) or NEMO knockout (grey bars) were used. Values were presented as mean ± SEM of total secretion in pg/ml,
nd means non-detectable. Comparison was made with the two-tailed t-test. c. In addition to IL-6 murine IL-8
homologues KC, LIX and MIP-2 were used to further show activation of SASP signaling. mRNA of all three
homologues was measured by RT-qPCR in untreated and etoposide-treated MDFs (n = 5). Cells with wildtype
NEMO (black bars) or NEMO knockout (grey bars) were used. Values were presented as mean ± SEM of fold
change. Comparison was made with the two-tailed t-test. d. IL-8 homologue secretion was measured by ELISA in
conditioned media as previously described (n = 5). Values were presented as mean ± SEM of total secretion in pg/
ml, nd means non-detectable. Comparison was made with the two-tailed t-test.
https://doi.org/10.1371/journal.pcbi.1005741.g009
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
18 / 30
Methods
Mice experiments
Murine dermal fibroblasts from an inducible connective tissue-specific NEMO-deficient
mouse model were used for in-vitro experiments. This mouse line (Col(I)α2-CreERT+;
NEMOf/f) was generated by crossing Col(I)α2-CreERT transgenic mice [51] with NEMO
floxed mice [35]. These mice were backcrossed to C57BL/6J for at least 6 generations. They
were maintained in the Animal Facility of the University of Ulm with 12 h light–dark cycle
and SPF conditions. The breeding of the mice and all experiments were approved by the ani-
mal ethical committee (approval number, Tierversuch-Nr. 1102, Regierungspra¨sidium
Tu¨bingen, Germany). For mice genotyping standard PCR techniques were used. The
sequences of the primers used in this manuscript are summarized in S1 Table. Briefly, DNA
was isolated from the tail tip of an individual mouse using a commercial kit (Easy DNA kit,
Invitrogen). Purified DNA was later dissolved in TE and used for PCR amplification. The PCR
products were run in QIAxcel Advance system (Qiagen) using the program AM320 and then
documented digitally.
Isolation and culture of murine dermal fibroblasts
Murine dermal fibroblasts (MDFs) were isolated from ear skin of young mice and cultured as
previously described [52].
Induction of DNA damage
DNA damage was induced by adding etoposide to cell culture media at a concentration of
25 μM for 3 hours after overnight serum-starvation. Supernatants subsequently removed and
cells were rinsed with PBS before adding fresh culture media. Cells and/or media were used 24
h later for further analysis.
Cloning
Recombineering technology [53] was used to constract plasmids containing CDS of both Cre
recombinase and fluorescence reporter, mRuby2 or only mRuby2. pCAG-Cre vector (a gift
from Connie Cepko, Addgene plasmid # 13775) was used for the recombineering. In the first
construct, the aim was to insert the T2A-mRuby2 sequence before the stop codon of Cre
recombinase and in the second construct, the aim was to replace the Cre ORF with mRuby2
ORF. In brief, synthetic DNA fragments were synthesized either as gBlock (IDT) or as Gen-
eArt string (Thermo Scientific). Four DNA fragments were synthesized, the first one contained
5’ 50 bp homology regions to the vector (targeting 50 nucleotide upstream of Cre ORF stop
codon), chloramphenicol and ccdB cassettes and 3’ terminal 50 bp homology regions to the
vector (targeting 50 nucleotide downstream of last amino acid coding codon of Cre ORF, i.e.,
condon preceding the Cre ORF stop codon). The second synthetic fragment contained 5’ 50
bp homology regions to the vector (targeting 50 nucleotide upstream of Cre ORF start codon),
chloramphenicol and ccdB cassettes and 3’ terminal 50 bp homology regions to the vector (tar-
geting 50 nucleotide downsteram of Cre ORF stop codon). The third synthetic fragment con-
tained 5’ 50 bp homology regions to the vector (same as fragment 1), T2A sequence-mRuby2
ORF and 3’ terminal 50 bp homology regions to the vector (same as fragment 1). The fourth
synthetic fragment contained 5’ 50 bp homology regions to the vector (same as fragment 2),
mRuby2 ORF and 3’ terminal 50 bp homology regions to the vector (same as fragment 2). E.
coli containing pCAG-Cre was processed for electrocompetent using standard methods and
these electrocompetent E coli, containing pCAG-Cre were electroporated with a dual inducible
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
19 / 30
expression plasmid pSC101-ccdA-gbaA (a gift from Prof. A. Francis Stewart) and selected for
ampicillin 100μg/ml and tetracycline 3.5μg/ml at 30˚C. Next day, 4–5 colonies were expanded
and the expression of recombineering proteins, λphage redα, redβ and redγ and recA (redgbaA)
was induced by L-rhamnose (1.4mg/ml). After 1 h of L-rhamnose treatment, the induced E.
coli were processed for electrocompetent and then electroprorated either with synthetic DNA
fragments 1 or 2. After 1 h of recovery in SOC medium, the electroporated E coli, were plated
in LB-agar containing ampicillin 100μg/ml, tetracycline 3.5μg/ml, chloramphenicol 25μg/ml
and 1.4mg/ml L-arabinose. L-arabinose addition induced the expression of ccdA, the antidote
of ccdB in that only recombined plasmid containing E. coli can survive. Thereafter colonies
from fragment 1 and fragment 2 electroporated E. coli plates were picked and expanded for
the verification of first recombinant product using restriction digestion analyses. The corre-
sponding colony was expanded and redgbaA expression was induced by L-rhamnose for 1 h.
The induced E. coli containing either recombined DNA fragment 1 or fragment 2 were made
electrocompetent for the second round of recombineering. The E. coli, containing recombined
DNA fragment 1 then electroporated with synthetic DNA fragment 3. The E. coli, containing
recombined DNA fragment 2 were electroporated with synthetic DNA fragment 4. The recov-
ered electroporated E. coli were plated in LB-agar containing ampicillin 100μg/ml and incu-
bated at 37˚C overnight. Colonies from both plates were picked, expanded and verified for the
second recombinant products. The correct plasmids were sequenced and verified through
commercial services (Sequiserve, Germany). Plasmid preparation was performed using a com-
mercially available kit (Qiagen plasmid plus kit, Qiagen). This plasmid (pCAG-Cre-T2A-
mRuby2) can be obtained from the authors on request and was deposited in the Addgene
repository (Accession ID 102989).
Initiation of Cre activity (NEMO knockout)
Early passage MDFs with a floxed NEMO allele were transfected with a Cre expressing vector
using an electroporation-based transfection method (Amaxa, Lonza Group). Transfer of the
plasmid was performed using a commercial kit with the AMAXA program N24 (Nucleofector
Kits for Mouse or Rat Hepatocytes, Lonza). Successful NEMO knockout was assessed by PCR
as explained before.
FACS sorting of positive cells
Two days after transfection cell populations were purified using the mRuby2-based reporter
system included in the previously described Cre-expressing vectors. Gating was set for living
cells and singlets, sorting was based on mRuby2 expression in the PE-channel. FACS-sorting
was performed with a FACSAria III system (BD Biosciences) and analysis was done on FACS-
Diva and FlowJo (Tree Star) software.
Immunofluorescence staining
Cells were fixed in 4% PFA in PBS for 15 min and thereafter treated with 0.1% Triton X-100
for 10 min at room temperature. Blocking was performed in 5% BSA for 1 h at room tempera-
ture. Anti-p65 (#8242, 1:200, Cell Signaling) and anti-γH2A.x (ab22551, 1:200, Abcam) were
used as primary antibodies overnight at 4˚C. Incubation with the secondary antibody Alexa
488 goat anti-mouse (for γH2A.x, 1:500) and Alexa 555 goat anti-rabbit (for p65, 1:500) was
performed at room temperature for 1 h.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
20 / 30
Western blotting
Western blot analyses were performed as described earlier [54]. In brief, murine dermal
fibroblasts were lysed in RIPA lysis buffer (25mM Tris-HCl pH 7.6, 150mM NaCl, 1% NP-
40, 1% sodium deoxycholate, 0.1% SDS) supplemented with protease and phosphatase
inhibitors (Thermo Scientific). Cells in RIPA were sonicated using sonopuls HD 2070 and
MS72 microtips (Bandelin). The sonicator setting was 50% power 3 cycles and 10 sec for
three times. Following sonication, the lysate was centrifuged for 15 min at 14000 rpm and
4˚C. The supernatant was collected and protein concentration was measured by Bradford
Assay (Biorad). 50μg of protein from each lysate was resolved in 4–20% SDS-PAGE, fol-
lowed by transfer to nitrocellulose membrane and probing the membrane with anti-NEMO
antibody (1:1000, Abcam). The membrane was incubated with goat anti-rabbit IgG coupled
with HRP for 1 hr (Jackson ImmunoResearch). Thereafter the membrane was developed by
LumiGLO chemiluminescence reagent (Cell Signaling Technologies) using Fusion FX7 Gel-
doc system (Vilber Lourmat), followed by stripping with Restore Plus Western blot Strip-
ping Buffer (Thermo Scientific) and re-probed with anti-β-actin antibody coupled with
HRP (1:12000, Santa Cruz), finally developed the membrane using LumiGLO.
Quantitative PCR
Twenty-four hours after treatment, total RNA was isolated from cultured murine dermal
fibroblasts using a commercial kit (RNeasy Mini Kit, Qiagen) as described by the manufac-
turer. Two μg of RNA per sample were reverse transcribed using illustra Ready-To-Go
RT-PCR Beads (GE Healthcare). Quantity and quality of total RNA and cDNA was assessed
using Nanodrop 1000 (Thermo Scientific) and QIAxcel Advance system (Qiagen). The 7300
real time PCR system (Applied Biosystem, Life Technologies) was used to amplify cDNA using
Power SYBR green mastermix (Applied Biosystems, Life Technologies). Sequences for primers
used in all experiments and genotyping are provided in S1 Table.
ELISA
After etoposide treatment cells were supplied with fresh culture media. Culture media was
taken for analysis of secreted IL-6 and murine IL-8 homologues (KC and MIP-2) 24 h after
treatment. Media was stored at -80˚C until analysis.
Concentrations of secreted IL-6 and murine IL-8 homologues after DNA damage were
determined using commercial kits (Mouse IL-6/KC/MIP-2 Quantikine ELISA Kit, R&D) as
described by the manufacturer.
Statistical calculations
The influence of a NEMO knockout was compared to wildtype controls based on IL-6, IL-8
homologue and p21 mRNA expression as well as IL-6 and IL-8 homologue protein secretion.
The sample size for all experiments was 5 per group. The expression and secretion of the two
groups was tested using unpaired two-tailed t-test. Furthermore, the influence of the NEMO
knockout compared to wildtype controls on the nuclear translocation of p65 was measured
by the percentage of fluorescence intensity in the cell nucleus as well as cytoplasm (sample
size = 10). The fluorescence intensity was tested using unpaired two-tailed t-test. The exact
p-values are depicted in the respective figures. The figures show mean values. Error bars corre-
spond to the standard error of the mean.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
21 / 30
Boolean networks
In a first step, IL- and DNA-damage pathways included in the Boolean model of SASP were
reconstructed individually. To generate the independent gene regulatory networks of inflam-
matory and DDR signaling, we collected peer-reviewed literature that is considered relevant in
the context of SASP (see Table 1). This literature reports data about the local interaction of key
genes regulating each pathway. The information was collected in murine and human experi-
mental in-vivo and in-vitro studies. In order to control the complexity of model we restricted
the set of regulatory factors in the model to the most relevant for SASP and to those being
important components of each pathway. The modeled pathways were chosen based on the
requirement in the onset and maintenance of the SASP shown in studies related to senescence
and the SASP. In total 80 publications were used to determine the relationships between the
individual components of the model (Table 1).
After the reconstruction of Boolean network models of inflammation and DNA damage
response, both were combined into a larger network. The impact of combining the two net-
work models instead of simulating them independently is shown by additional analysis in S1
Text. Simulations based on specific environmental (input) conditions were performed to find
the corresponding attractors. Furthermore, to identify possible interaction targets, i.e., to gen-
erate testable hypotheses about interventions, we fixed corresponding regulatory factors to
either 0 or 1 (modelling of knockout or overexpression, similar to [55]) and reran the simula-
tions (S1 Text). Given an interaction target, we looked for the attractors that positively influ-
ence the DNA damage response phenotype.
Network figures were drawn with Biotapestry (www.biotapestry.org). Simulations of the
Boolean network were performed with the package BoolNet [12, 56] in R (www.r-project.org).
This model contains two external signals (DNA damage and Activated Oncogenes). These
signals do not coincide with genes within the network, but represent different stimuli from
external or internal sources that are known to activate the DNA damage response and/or cell
cycle arrest signaling through either p16INK4 or p53/p21.
Supporting information
S1 Fig. Establishment of a pure NEMO knockout murine dermal fibroblast (MDF) popula-
tion. a. To purify NEMO k/o MDFs, NEMO-floxed cells were transfected with a Cre-recombi-
nase vector including a mRUBY2-reporter construct. Two days post-transfection cells were
purified for the NEMO k/o using flowcytometry-based sorting, gating for living cells, cell sin-
glets and mRUBY2 signal (histograms; left to right). b. Successful NEMO k/o was determined
using PCR analysis. DNA was isolated from FACS-sorted MDFs and later used for PCR ampli-
fication. Cre-recombinase activity induced the deletion of floxed NEMO alleles resulting in a
bigger sized amplification product in successful knockouts as compared to wildtype cells. c. In
addition to PCR analysis a successful knockout on protein level was determined by western
blotting of cell lysates equilibrated to actin expression levels.
(TIF)
S2 Fig. Unaltered expression of selected genes (predicted to be unaffected in NEMO knock-
out) following NEMO knockout. The expression level of a set of genes that were predicted
not to be changed after NEMO knockout by the Boolean network model. In a setting of 2-fold
cutoff (blue dotted line), the expression of all genes remained unaltered between control and
NEMO knock out MDFs. Dotted line at value ‘1’ represents level of expression in the control
MDFs.
(TIF)
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
22 / 30
S1 Table. Primer sequences.
(DOCX)
S1 Text. Simulation of SASP network with BoolNet.
(PDF)
Author Contributions
Conceptualization: Patrick Meyer, Pallab Maity, Andre Burkovski, Christoph Mu¨ssel, Hans
A. Kestler, Karin Scharffetter-Kochanek.
Formal analysis: Hans A. Kestler.
Funding acquisition: Hans A. Kestler, Karin Scharffetter-Kochanek.
Investigation: Patrick Meyer, Pallab Maity, Julian Schwab, Hans A. Kestler, Karin Scharffet-
ter-Kochanek.
Methodology: Andre Burkovski, Karmveer Singh, Filipa F. Ferreira, Linda Krug, Meinhard
Wlaschek, Hans A. Kestler.
Project administration: Meinhard Wlaschek, Hans A. Kestler, Karin Scharffetter-Kochanek.
Resources: Harald J. Maier, Thomas Wirth, Karin Scharffetter-Kochanek.
Software: Andre Burkovski, Julian Schwab, Christoph Mu¨ssel, Hans A. Kestler.
Supervision: Hans A. Kestler, Karin Scharffetter-Kochanek.
Validation: Harald J. Maier, Thomas Wirth.
Writing – original draft: Patrick Meyer, Andre Burkovski, Hans A. Kestler, Karin Scharffet-
ter-Kochanek.
Writing – review & editing: Pallab Maity, Julian Schwab, Christoph Mu¨ssel, Hans A. Kestler,
Karin Scharffetter-Kochanek.
References
1.
Goldman DP, Cutler D, Rowe JW, Michaud PC, Sullivan J, Peneva D, et al. Substantial health and
economic returns from delayed aging may warrant a new focus for medical research. Health Aff (Mill-
wood). 2013; 32(10):1698–705. https://doi.org/10.1377/hlthaff.2013.0052 PMID: 24101058; PubMed
Central PMCID: PMC3938188.
2.
Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, et al. Senescence and
tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007; 445
(7128):656–60. https://doi.org/10.1038/nature05529 PMID: 17251933.
3.
Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, et al. Oncogene-
induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008; 133
(6):1019–31. https://doi.org/10.1016/j.cell.2008.03.039 PMID: 18555778.
4.
Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, et al. Senescence-associated secretory
phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor.
PLoS Biol. 2008; 6(12):2853–68. https://doi.org/10.1371/journal.pbio.0060301 PMID: 19053174;
PubMed Central PMCID: PMC2592359.
5.
Coppe JP, Patil CK, Rodier F, Krtolica A, Beausejour CM, Parrinello S, et al. A human-like senes-
cence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxy-
gen. PloS one. 2010; 5(2):e9188. https://doi.org/10.1371/journal.pone.0009188 PMID: 20169192;
PubMed Central PMCID: PMC2820538.
6.
Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, et al. A complex secretory pro-
gram orchestrated by the inflammasome controls paracrine senescence. Nature cell biology. 2013; 15
(8):978–90. https://doi.org/10.1038/ncb2784 PMID: 23770676; PubMed Central PMCID:
PMCPMC3732483.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
23 / 30
7.
Acosta JC, O’Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, et al. Chemokine signaling via the
CXCR2 receptor reinforces senescence. Cell. 2008; 133(6):1006–18. https://doi.org/10.1016/j.cell.
2008.03.038 PMID: 18555777.
8.
Cohen HJ, Pieper CF, Harris T, Rao KM, Currie MS. The association of plasma IL-6 levels with func-
tional disability in community-dwelling elderly. J Gerontol A Biol Sci Med Sci. 1997; 52(4):M201–8.
PMID: 9224431.
9.
Rodier F, Coppe JP, Patil CK, Hoeijmakers WA, Munoz DP, Raza SR, et al. Persistent DNA damage
signalling triggers senescence-associated inflammatory cytokine secretion. Nature cell biology. 2009;
11(8):973–9. https://doi.org/10.1038/ncb1909 PMID: 19597488; PubMed Central PMCID:
PMC2743561.
10.
Davidson EH, Erwin DH. Gene regulatory networks and the evolution of animal body plans. Science.
2006; 311(5762):796–800. https://doi.org/10.1126/science.1113832 PMID: 16469913.
11.
Kestler HA, Wawra C, Kracher B, Ku¨hl M. Network modeling of signal transduction: establishing the
global view. Bioessays. 2008; 30(11–12):1110–25. https://doi.org/10.1002/bies.20834 PMID:
18937364.
12.
Naldi A, Monteiro PT, Mu¨ssel C, Consortium for Logical M, Tools, Kestler HA, et al. Cooperative devel-
opment of logical modelling standards and tools with CoLoMoTo. Bioinformatics. 2015; 31(7):1154–9.
https://doi.org/10.1093/bioinformatics/btv013 PMID: 25619997.
13.
Shmulevich I, Dougherty ER. Genomic Signal Processing: Princeton University Press; 2007.
14.
de Jong H. Modeling and simulation of genetic regulatory systems: a literature review. J Comput Biol.
2002; 9(1):67–103. https://doi.org/10.1089/10665270252833208 PMID: 11911796.
15.
Kauffman S. Homeostasis and differentiation in random genetic control networks. Nature. 1969; 224
(5215):177–8. PMID: 5343519.
16.
Le Novere N. Quantitative and logic modelling of molecular and gene networks. Nat Rev Genet. 2015;
16(3):146–58. https://doi.org/10.1038/nrg3885 PMID: 25645874; PubMed Central PMCID:
PMCPMC4604653.
17.
Fisher J, Henzinger TA. Executable cell biology. Nature biotechnology. 2007; 25(11):1239–49. https://
doi.org/10.1038/nbt1356 PMID: 17989686.
18.
Harvey I, Bossomaier T. Time Out of Joint: Attractors in Asynchronous Ramdom Boolean Networks.
In: Husbands P, Harvey I, editors. Proceedings of the Fourth European Artificial Life Conference: MIT
Press; 1997. p. 67–75.
19.
Klarner H, Bockmayr A, Siebert H. Computing Symbolic Steady States of Boolean Networks. In: Wąs
J, Sirakoulis GC, Bandini S, editors. Cellular Automata: 11th International Conference on Cellular
Automata for Research and Industry, ACRI 2014, Krakow, Poland, September 22–25, 2014 Proceed-
ings. Cham: Springer International Publishing; 2014. p. 561–70.
20.
Zañudo JGT, Albert R. An effective network reduction approach to find the dynamical repertoire of dis-
crete dynamic networks. Chaos: An Interdisciplinary Journal of Nonlinear Science. 2013; 23
(2):025111. https://doi.org/10.1063/1.4809777 PMID: 23822509
21.
Dahlhaus M, Burkovski A, Hertwig F, Mussel C, Volland R, Fischer M, et al. Boolean modeling identi-
fies Greatwall/MASTL as an important regulator in the AURKA network of neuroblastoma. Cancer let-
ters. 2016; 371(1):79–89. https://doi.org/10.1016/j.canlet.2015.11.025 PMID: 26616283.
22.
MacLean D, Studholme DJ. A Boolean model of the Pseudomonas syringae hrp regulon predicts a
tightly regulated system. PloS one. 2010; 5(2):e9101. https://doi.org/10.1371/journal.pone.0009101
PMID: 20169167; PubMed Central PMCID: PMC2821412.
23.
Moignard V, Woodhouse S, Haghverdi L, Lilly AJ, Tanaka Y, Wilkinson AC, et al. Decoding the regula-
tory network of early blood development from single-cell gene expression measurements. Nature bio-
technology. 2015; 33(3):269–76. https://doi.org/10.1038/nbt.3154 PMID: 25664528; PubMed Central
PMCID: PMCPMC4374163.
24.
Herrmann F, Gross A, Zhou D, Kestler HA, Ku¨hl M. A boolean model of the cardiac gene regulatory
network determining first and second heart field identity. PloS one. 2012; 7(10):e46798. https://doi.
org/10.1371/journal.pone.0046798 PMID: 23056457; PubMed Central PMCID: PMC3462786.
25.
Kestler HA, Kuhl M. From individual Wnt pathways towards a Wnt signalling network. Philosophical
transactions of the Royal Society of London Series B, Biological sciences. 2008; 363(1495):1333–47.
https://doi.org/10.1098/rstb.2007.2251 PMID: 18192173; PubMed Central PMCID:
PMCPMC2610122.
26.
Feist AM, Herrgard MJ, Thiele I, Reed JL, Palsson BO. Reconstruction of biochemical networks in
microorganisms. Nat Rev Microbiol. 2009; 7(2):129–43. https://doi.org/10.1038/nrmicro1949 PMID:
19116616; PubMed Central PMCID: PMCPMC3119670.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
24 / 30
27.
Saadatpour A, Albert R, Reluga TC. A Reduction Method for Boolean Network Models Proven to Con-
serve Attractors. SIAM Journal on Applied Dynamical Systems. 2013; 12(4):1997–2011. https://doi.
org/10.1137/13090537x
28.
Veliz-Cuba A. Reduction of Boolean network models. Journal of theoretical biology. 2011; 289:167–
72. https://doi.org/10.1016/j.jtbi.2011.08.042 PMID: 21907211.
29.
Hiscott J, Marois J, Garoufalis J, D’Addario M, Roulston A, Kwan I, et al. Characterization of a func-
tional NF-kappa B site in the human interleukin 1 beta promoter: evidence for a positive autoregulatory
loop. Molecular and cellular biology. 1993; 13(10):6231–40. PMID: 8413223; PubMed Central PMCID:
PMC364682.
30.
Mori N, Prager D. Transactivation of the interleukin-1alpha promoter by human T-cell leukemia virus
type I and type II Tax proteins. Blood. 1996; 87(8):3410–7. PMID: 8605359.
31.
Holtmann H, Winzen R, Holland P, Eickemeier S, Hoffmann E, Wallach D, et al. Induction of interleu-
kin-8 synthesis integrates effects on transcription and mRNA degradation from at least three different
cytokine- or stress-activated signal transduction pathways. Molecular and cellular biology. 1999; 19
(10):6742–53. PMID: 10490613; PubMed Central PMCID: PMC84667.
32.
Libermann TA, Baltimore D. Activation of interleukin-6 gene expression through the NF-kappa B tran-
scription factor. Molecular and cellular biology. 1990; 10(5):2327–34. PMID: 2183031; PubMed Cen-
tral PMCID: PMC360580.
33.
Son YH, Jeong YT, Lee KA, Choi KH, Kim SM, Rhim BY, et al. Roles of MAPK and NF-kappaB in inter-
leukin-6 induction by lipopolysaccharide in vascular smooth muscle cells. Journal of cardiovascular
pharmacology. 2008; 51(1):71–7. https://doi.org/10.1097/FJC.0b013e31815bd23d PMID: 18209571.
34.
Park GY, Le S, Park KH, Le CT, Kim YW, Han SK, et al. Anti-inflammatory effect of adenovirus-medi-
ated IkappaBalpha overexpression in respiratory epithelial cells. The European respiratory journal.
2001; 18(5):801–9. PMID: 11757631.
35.
Schmidt-Supprian M, Bloch W, Courtois G, Addicks K, Israel A, Rajewsky K, et al. NEMO/IKK
gamma-deficient mice model incontinentia pigmenti. Molecular cell. 2000; 5(6):981–92. PMID:
10911992.
36.
Salminen A, Suuronen T, Huuskonen J, Kaarniranta K. NEMO shuttle: a link between DNA damage
and NF-kappaB activation in progeroid syndromes? Biochemical and biophysical research communi-
cations. 2008; 367(4):715–8. https://doi.org/10.1016/j.bbrc.2007.11.189 PMID: 18201555.
37.
Zurgil U, Ben-Ari A, Atias K, Isakov N, Apte R, Livneh E. PKCeta promotes senescence induced by
oxidative stress and chemotherapy. Cell death & disease. 2014; 5:e1531. https://doi.org/10.1038/
cddis.2014.481 PMID: 25412309; PubMed Central PMCID: PMCPMC4260739.
38.
Dyson N. The regulation of E2F by pRB-family proteins. Genes & development. 1998; 12(15):2245–
62. PMID: 9694791.
39.
Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-kappaB signaling in the induction of
senescence-associated secretory phenotype (SASP). Cellular signalling. 2012; 24(4):835–45. https://
doi.org/10.1016/j.cellsig.2011.12.006 PMID: 22182507.
40.
Sebban H, Yamaoka S, Courtois G. Posttranslational modifications of NEMO and its partners in NF-
kappaB signaling. Trends Cell Biol. 2006; 16(11):569–77. https://doi.org/10.1016/j.tcb.2006.09.004
PMID: 16987664.
41.
Israel A. The IKK complex, a central regulator of NF-kappaB activation. Cold Spring Harbor perspec-
tives in biology. 2010; 2(3):a000158. https://doi.org/10.1101/cshperspect.a000158 PMID: 20300203;
PubMed Central PMCID: PMCPMC2829958.
42.
Di Micco R, Fumagalli M, Cicalese A, Piccinin S, Gasparini P, Luise C, et al. Oncogene-induced
senescence is a DNA damage response triggered by DNA hyper-replication. Nature. 2006; 444
(7119):638–42. https://doi.org/10.1038/nature05327 PMID: 17136094.
43.
Gensler HL, Bernstein H. DNA damage as the primary cause of aging. Q Rev Biol. 1981; 56(3):279–
303. PMID: 7031747.
44.
Holmes GE, Bernstein C, Bernstein H. Oxidative and other DNA damages as the basis of aging: a
review. Mutat Res. 1992; 275(3–6):305–15. PMID: 1383772.
45.
Badache A, Hynes NE. Interleukin 6 inhibits proliferation and, in cooperation with an epidermal growth
factor receptor autocrine loop, increases migration of T47D breast cancer cells. Cancer research.
2001; 61(1):383–91. PMID: 11196191.
46.
Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the
dark side of tumor suppression. Annu Rev Pathol. 2010; 5:99–118. https://doi.org/10.1146/annurev-
pathol-121808-102144 PMID: 20078217; PubMed Central PMCID: PMCPMC4166495.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
25 / 30
47.
Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, et al. Senescence of activated
stellate cells limits liver fibrosis. Cell. 2008; 134(4):657–67. https://doi.org/10.1016/j.cell.2008.06.049
PMID: 18724938; PubMed Central PMCID: PMCPMC3073300.
48.
Lee JK, Bettencourt R, Brenner D, Le TA, Barrett-Connor E, Loomba R. Association between serum
interleukin-6 concentrations and mortality in older adults: the Rancho Bernardo study. PloS one. 2012;
7(4):e34218. https://doi.org/10.1371/journal.pone.0034218 PMID: 22514624; PubMed Central
PMCID: PMCPMC3325993.
49.
Habineza Ndikuyeze G, Gaurnier-Hausser A, Patel R, Baldwin AS, May MJ, Flood P, et al. A phase I
clinical trial of systemically delivered NEMO binding domain peptide in dogs with spontaneous acti-
vated B-cell like diffuse large B-cell lymphoma. PloS one. 2014; 9(5):e95404. https://doi.org/10.1371/
journal.pone.0095404 PMID: 24798348; PubMed Central PMCID: PMCPMC4010398.
50.
Dai S, Hirayama T, Abbas S, Abu-Amer Y. The IkappaB kinase (IKK) inhibitor, NEMO-binding domain
peptide, blocks osteoclastogenesis and bone erosion in inflammatory arthritis. The Journal of biologi-
cal chemistry. 2004; 279(36):37219–22. https://doi.org/10.1074/jbc.C400258200 PMID: 15252035.
51.
Zheng B, Zhang Z, Black CM, de Crombrugghe B, Denton CP. Ligand-dependent genetic recombina-
tion in fibroblasts: a potentially powerful technique for investigating gene function in fibrosis. The Amer-
ican journal of pathology. 2002; 160(5):1609–17. https://doi.org/10.1016/S0002-9440(10)61108-X
PMID: 12000713; PubMed Central PMCID: PMC1850857.
52.
Treiber N, Maity P, Singh K, Kohn M, Keist AF, Ferchiu F, et al. Accelerated aging phenotype in mice
with conditional deficiency for mitochondrial superoxide dismutase in the connective tissue. Aging cell.
2011; 10(2):239–54. https://doi.org/10.1111/j.1474-9726.2010.00658.x PMID: 21108731.
53.
Wang H, Bian X, Xia L, Ding X, Muller R, Zhang Y, et al. Improved seamless mutagenesis by recombi-
neering using ccdB for counterselection. Nucleic acids research. 2014; 42(5):e37. https://doi.org/10.
1093/nar/gkt1339 PMID: 24369425; PubMed Central PMCID: PMCPMC3950717.
54.
Singh K, Maity P, Krug L, Meyer P, Treiber N, Lucas T, et al. Superoxide anion radicals induce IGF-1
resistance through concomitant activation of PTP1B and PTEN. EMBO Mol Med. 2015; 7(1):59–77.
https://doi.org/10.15252/emmm.201404082 PMID: 25520316; PubMed Central PMCID:
PMCPMC4309668.
55.
Zañudo JG, Albert R. Cell fate reprogramming by control of intracellular network dynamics. PLoS
computational biology. 2015; 11(4):e1004193. https://doi.org/10.1371/journal.pcbi.1004193 PMID:
25849586; PubMed Central PMCID: PMCPMC4388852.
56.
Mu¨ssel C, Hopfensitz M, Kestler HA. BoolNet—an R package for generation, reconstruction and anal-
ysis of Boolean networks. Bioinformatics. 2010; 26(10):1378–80. https://doi.org/10.1093/
bioinformatics/btq124 PMID: 20378558.
57.
Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K, Linn S. Molecular mechanisms of mammalian DNA
repair and the DNA damage checkpoints. Annual review of biochemistry. 2004; 73:39–85. https://doi.
org/10.1146/annurev.biochem.73.011303.073723 PMID: 15189136.
58.
Shiloh Y. ATM and related protein kinases: safeguarding genome integrity. Nature reviews Cancer.
2003; 3(3):155–68. https://doi.org/10.1038/nrc1011 PMID: 12612651.
59.
Smith J, Tho LM, Xu N, Gillespie DA. The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signal-
ing and cancer. Advances in cancer research. 2010; 108:73–112. https://doi.org/10.1016/B978-0-12-
380888-2.00003-0 PMID: 21034966.
60.
Falck J, Mailand N, Syljuasen RG, Bartek J, Lukas J. The ATM-Chk2-Cdc25A checkpoint pathway
guards against radioresistant DNA synthesis. Nature. 2001; 410(6830):842–7. https://doi.org/10.
1038/35071124 PMID: 11298456.
61.
Zhan Q, Fan S, Smith ML, Bae I, Yu K, Alamo I Jr., et al. Abrogation of p53 function affects gadd gene
responses to DNA base-damaging agents and starvation. DNA and cell biology. 1996; 15(10):805–15.
https://doi.org/10.1089/dna.1996.15.805 PMID: 8892753.
62.
Appella E, Anderson CW. Post-translational modifications and activation of p53 by genotoxic stresses.
European journal of biochemistry / FEBS. 2001; 268(10):2764–72. PMID: 11358490.
63.
Hirao A, Kong YY, Matsuoka S, Wakeham A, Ruland J, Yoshida H, et al. DNA damage-induced activa-
tion of p53 by the checkpoint kinase Chk2. Science. 2000; 287(5459):1824–7. PMID: 10710310.
64.
Cheng Q, Chen L, Li Z, Lane WS, Chen J. ATM activates p53 by regulating MDM2 oligomerization
and E3 processivity. The EMBO journal. 2009; 28(24):3857–67. https://doi.org/10.1038/emboj.2009.
294 PMID: 19816404; PubMed Central PMCID: PMC2797053.
65.
Jones SN, Roe AE, Donehower LA, Bradley A. Rescue of embryonic lethality in Mdm2-deficient mice
by absence of p53. Nature. 1995; 378(6553):206–8. https://doi.org/10.1038/378206a0 PMID:
7477327.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
26 / 30
66.
Ke Q, Costa M. Hypoxia-inducible factor-1 (HIF-1). Molecular pharmacology. 2006; 70(5):1469–80.
https://doi.org/10.1124/mol.106.027029 PMID: 16887934.
67.
Blagosklonny MV, An WG, Romanova LY, Trepel J, Fojo T, Neckers L. p53 inhibits hypoxia-inducible
factor-stimulated transcription. The Journal of biological chemistry. 1998; 273(20):11995–8. PMID:
9575138.
68.
Xiong Y, Hannon GJ, Zhang H, Casso D, Kobayashi R, Beach D. p21 is a universal inhibitor of cyclin
kinases. Nature. 1993; 366(6456):701–4. https://doi.org/10.1038/366701a0 PMID: 8259214.
69.
Herold S, Wanzel M, Beuger V, Frohme C, Beul D, Hillukkala T, et al. Negative regulation of the mam-
malian UV response by Myc through association with Miz-1. Molecular cell. 2002; 10(3):509–21.
PMID: 12408820.
70.
Brugarolas J, Moberg K, Boyd SD, Taya Y, Jacks T, Lees JA. Inhibition of cyclin-dependent kinase 2
by p21 is necessary for retinoblastoma protein-mediated G1 arrest after gamma-irradiation. Proceed-
ings of the National Academy of Sciences of the United States of America. 1999; 96(3):1002–7. PMID:
9927683; PubMed Central PMCID: PMC15340.
71.
Grafstrom RH, Pan W, Hoess RH. Defining the substrate specificity of cdk4 kinase-cyclin D1 complex.
Carcinogenesis. 1999; 20(2):193–8. PMID: 10069453.
72.
Weinberg RA. The retinoblastoma protein and cell cycle control. Cell. 1995; 81(3):323–30. PMID:
7736585.
73.
Barak Y, Juven T, Haffner R, Oren M. mdm2 expression is induced by wild type p53 activity. The
EMBO journal. 1993; 12(2):461–8. PMID: 8440237; PubMed Central PMCID: PMC413229.
74.
Moll UM, Petrenko O. The MDM2-p53 interaction. Molecular cancer research: MCR. 2003; 1
(14):1001–8. PMID: 14707283.
75.
Shapiro GI, Edwards CD, Ewen ME, Rollins BJ. p16INK4A participates in a G1 arrest checkpoint in
response to DNA damage. Molecular and cellular biology. 1998; 18(1):378–87. PMID: 9418885;
PubMed Central PMCID: PMC121508.
76.
Wuerzberger-Davis SM, Nakamura Y, Seufzer BJ, Miyamoto S. NF-kappaB activation by combina-
tions of NEMO SUMOylation and ATM activation stresses in the absence of DNA damage. Oncogene.
2007; 26(5):641–51. https://doi.org/10.1038/sj.onc.1209815 PMID: 16862178.
77.
Yang Y, Xia F, Hermance N, Mabb A, Simonson S, Morrissey S, et al. A cytosolic ATM/NEMO/RIP1
complex recruits TAK1 to mediate the NF-kappaB and p38 mitogen-activated protein kinase (MAPK)/
MAPK-activated protein 2 responses to DNA damage. Molecular and cellular biology. 2011; 31
(14):2774–86. https://doi.org/10.1128/MCB.01139-10 PMID: 21606198; PubMed Central PMCID:
PMC3133388.
78.
Wu ZH, Wong ET, Shi Y, Niu J, Chen Z, Miyamoto S, et al. ATM- and NEMO-dependent ELKS ubiqui-
tination coordinates TAK1-mediated IKK activation in response to genotoxic stress. Molecular cell.
2010; 40(1):75–86. https://doi.org/10.1016/j.molcel.2010.09.010 PMID: 20932476; PubMed Central
PMCID: PMC3048026.
79.
Ling L, Cao Z, Goeddel DV. NF-kappaB-inducing kinase activates IKK-alpha by phosphorylation of
Ser-176. Proceedings of the National Academy of Sciences of the United States of America. 1998; 95
(7):3792–7. PMID: 9520446; PubMed Central PMCID: PMC19916.
80.
Bai D, Ueno L, Vogt PK. Akt-mediated regulation of NFkappaB and the essentialness of NFkappaB for
the oncogenicity of PI3K and Akt. International journal of cancer Journal international du cancer. 2009;
125(12):2863–70. https://doi.org/10.1002/ijc.24748 PMID: 19609947; PubMed Central PMCID:
PMCPMC2767458.
81.
Sun SC, Ganchi PA, Ballard DW, Greene WC. NF-kappa B controls expression of inhibitor I kappa B
alpha: evidence for an inducible autoregulatory pathway. Science. 1993; 259(5103):1912–5. PMID:
8096091.
82.
Zandi E, Rothwarf DM, Delhase M, Hayakawa M, Karin M. The IkappaB kinase complex (IKK) con-
tains two kinase subunits, IKKalpha and IKKbeta, necessary for IkappaB phosphorylation and NF-kap-
paB activation. Cell. 1997; 91(2):243–52. PMID: 9346241.
83.
Hinz M, Scheidereit C. The IkappaB kinase complex in NF-kappaB regulation and beyond. EMBO
reports. 2014; 15(1):46–61. https://doi.org/10.1002/embr.201337983 PMID: 24375677; PubMed Cen-
tral PMCID: PMCPMC4303448.
84.
Weber A, Wasiliew P, Kracht M. Interleukin-1 (IL-1) pathway. Science signaling. 2010; 3(105):cm1.
https://doi.org/10.1126/scisignal.3105cm1 PMID: 20086235.
85.
Kawagoe T, Sato S, Matsushita K, Kato H, Matsui K, Kumagai Y, et al. Sequential control of Toll-like
receptor-dependent responses by IRAK1 and IRAK2. Nature immunology. 2008; 9(6):684–91. https://
doi.org/10.1038/ni.1606 PMID: 18438411.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
27 / 30
86.
Li S, Strelow A, Fontana EJ, Wesche H. IRAK-4: a novel member of the IRAK family with the proper-
ties of an IRAK-kinase. Proceedings of the National Academy of Sciences of the United States of
America. 2002; 99(8):5567–72. https://doi.org/10.1073/pnas.082100399 PMID: 11960013; PubMed
Central PMCID: PMC122810.
87.
Wesche H, Henzel WJ, Shillinglaw W, Li S, Cao Z. MyD88: an adapter that recruits IRAK to the IL-1
receptor complex. Immunity. 1997; 7(6):837–47. PMID: 9430229.
88.
Yamasaki K, Taga T, Hirata Y, Yawata H, Kawanishi Y, Seed B, et al. Cloning and expression of the
human interleukin-6 (BSF-2/IFN beta 2) receptor. Science. 1988; 241(4867):825–8. PMID: 3136546.
89.
Yao J, Kim TW, Qin J, Jiang Z, Qian Y, Xiao H, et al. Interleukin-1 (IL-1)-induced TAK1-dependent
Versus MEKK3-dependent NFkappaB activation pathways bifurcate at IL-1 receptor-associated
kinase modification. The Journal of biological chemistry. 2007; 282(9):6075–89. https://doi.org/10.
1074/jbc.M609039200 PMID: 17197697.
90.
Kanayama A, Seth RB, Sun L, Ea CK, Hong M, Shaito A, et al. TAB2 and TAB3 activate the NF-kap-
paB pathway through binding to polyubiquitin chains. Molecular cell. 2004; 15(4):535–48. https://doi.
org/10.1016/j.molcel.2004.08.008 PMID: 15327770.
91.
Ninomiya-Tsuji J, Kishimoto K, Hiyama A, Inoue J, Cao Z, Matsumoto K. The kinase TAK1 can acti-
vate the NIK-I kappaB as well as the MAP kinase cascade in the IL-1 signalling pathway. Nature.
1999; 398(6724):252–6. https://doi.org/10.1038/18465 PMID: 10094049.
92.
Wang C, Deng L, Hong M, Akkaraju GR, Inoue J, Chen ZJ. TAK1 is a ubiquitin-dependent kinase of
MKK and IKK. Nature. 2001; 412(6844):346–51. https://doi.org/10.1038/35085597 PMID: 11460167.
93.
Hammaker DR, Boyle DL, Chabaud-Riou M, Firestein GS. Regulation of c-Jun N-terminal kinase by
MEKK-2 and mitogen-activated protein kinase kinase kinases in rheumatoid arthritis. Journal of immu-
nology. 2004; 172(3):1612–8. PMID: 14734742.
94.
Finch A, Holland P, Cooper J, Saklatvala J, Kracht M. Selective activation of JNK/SAPK by interleukin-
1 in rabbit liver is mediated by MKK7. FEBS letters. 1997; 418(1–2):144–8. PMID: 9414114.
95.
Tournier C, Dong C, Turner TK, Jones SN, Flavell RA, Davis RJ. MKK7 is an essential component of
the JNK signal transduction pathway activated by proinflammatory cytokines. Genes & development.
2001; 15(11):1419–26. https://doi.org/10.1101/gad.888501 PMID: 11390361; PubMed Central
PMCID: PMC312702.
96.
Kristiansen M, Hughes R, Patel P, Jacques TS, Clark AR, Ham J. Mkp1 is a c-Jun target gene that
antagonizes JNK-dependent apoptosis in sympathetic neurons. The Journal of neuroscience: the offi-
cial journal of the Society for Neuroscience. 2010; 30(32):10820–32. https://doi.org/10.1523/
JNEUROSCI.2824-10.2010 PMID: 20702711; PubMed Central PMCID: PMC3044878.
97.
Inoue T, Hammaker D, Boyle DL, Firestein GS. Regulation of p38 MAPK by MAPK kinases 3 and 6 in
fibroblast-like synoviocytes. Journal of immunology. 2005; 174(7):4301–6. PMID: 15778394.
98.
Kiemer AK, Weber NC, Furst R, Bildner N, Kulhanek-Heinze S, Vollmar AM. Inhibition of p38 MAPK
activation via induction of MKP-1: atrial natriuretic peptide reduces TNF-alpha-induced actin polymeri-
zation and endothelial permeability. Circulation research. 2002; 90(8):874–81. PMID: 11988488.
99.
van Dam H, Wilhelm D, Herr I, Steffen A, Herrlich P, Angel P. ATF-2 is preferentially activated by
stress-activated protein kinases to mediate c-jun induction in response to genotoxic agents. The
EMBO journal. 1995; 14(8):1798–811. PMID: 7737130; PubMed Central PMCID: PMC398273.
100.
Wadgaonkar R, Pierce JW, Somnay K, Damico RL, Crow MT, Collins T, et al. Regulation of c-Jun N-
terminal kinase and p38 kinase pathways in endothelial cells. American journal of respiratory cell and
molecular biology. 2004; 31(4):423–31. https://doi.org/10.1165/rcmb.2003-0384OC PMID: 15231489.
101.
Minden A, Lin A, Smeal T, Derijard B, Cobb M, Davis R, et al. c-Jun N-terminal phosphorylation corre-
lates with activation of the JNK subgroup but not the ERK subgroup of mitogen-activated protein
kinases. Molecular and cellular biology. 1994; 14(10):6683–8. PMID: 7935387; PubMed Central
PMCID: PMC359198.
102.
Pulverer BJ, Kyriakis JM, Avruch J, Nikolakaki E, Woodgett JR. Phosphorylation of c-jun mediated by
MAP kinases. Nature. 1991; 353(6345):670–4. https://doi.org/10.1038/353670a0 PMID: 1922387.
103.
Hanlon M, Bundy LM, Sealy L. C/EBP beta and Elk-1 synergistically transactivate the c-fos serum
response element. BMC cell biology. 2000; 1:2. https://doi.org/10.1186/1471-2121-1-2 PMID:
11151091; PubMed Central PMCID: PMC29063.
104.
Silvers AL, Bachelor MA, Bowden GT. The role of JNK and p38 MAPK activities in UVA-induced sig-
naling pathways leading to AP-1 activation and c-Fos expression. Neoplasia. 2003; 5(4):319–29.
https://doi.org/10.1016/S1476-5586(03)80025-8 PMID: 14511403; PubMed Central PMCID:
PMC1502419.
105.
Babu GJ, Lalli MJ, Sussman MA, Sadoshima J, Periasamy M. Phosphorylation of elk-1 by MEK/ERK
pathway is necessary for c-fos gene activation during cardiac myocyte hypertrophy. Journal of
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
28 / 30
molecular and cellular cardiology. 2000; 32(8):1447–57. https://doi.org/10.1006/jmcc.2000.1185
PMID: 10900171.
106.
Cavigelli M, Dolfi F, Claret FX, Karin M. Induction of c-fos expression through JNK-mediated TCF/Elk-
1 phosphorylation. The EMBO journal. 1995; 14(23):5957–64. PMID: 8846788; PubMed Central
PMCID: PMC394715.
107.
Yang E, Lerner L, Besser D, Darnell JE Jr. Independent and cooperative activation of chromosomal c-
fos promoter by STAT3. The Journal of biological chemistry. 2003; 278(18):15794–9. https://doi.org/
10.1074/jbc.M213073200 PMID: 12600988.
108.
Angel P, Karin M. The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation.
Biochimica et biophysica acta. 1991; 1072(2–3):129–57. PMID: 1751545.
109.
Casals-Casas C, Alvarez E, Serra M, de la Torre C, Farrera C, Sanchez-Tillo E, et al. CREB and AP-1
activation regulates MKP-1 induction by LPS or M-CSF and their kinetics correlate with macrophage
activation versus proliferation. European journal of immunology. 2009; 39(7):1902–13. https://doi.org/
10.1002/eji.200839037 PMID: 19585511.
110.
Sanchez-Tillo E, Comalada M, Xaus J, Farrera C, Valledor AF, Caelles C, et al. JNK1 Is required for
the induction of Mkp1 expression in macrophages during proliferation and lipopolysaccharide-depen-
dent activation. The Journal of biological chemistry. 2007; 282(17):12566–73. https://doi.org/10.1074/
jbc.M609662200 PMID: 17337450.
111.
Kang HB, Kim YE, Kwon HJ, Sok DE, Lee Y. Enhancement of NF-kappaB expression and activity
upon differentiation of human embryonic stem cell line SNUhES3. Stem cells and development. 2007;
16(4):615–23. https://doi.org/10.1089/scd.2007.0014 PMID: 17784835.
112.
Kunsch C, Rosen CA. NF-kappa B subunit-specific regulation of the interleukin-8 promoter. Molecular
and cellular biology. 1993; 13(10):6137–46. PMID: 8413215; PubMed Central PMCID: PMC364673.
113.
Neff L, Zeisel M, Druet V, Takeda K, Klein JP, Sibilia J, et al. ERK 1/2- and JNKs-dependent synthesis
of interleukins 6 and 8 by fibroblast-like synoviocytes stimulated with protein I/II, a modulin from oral
streptococci, requires focal adhesion kinase. The Journal of biological chemistry. 2003; 278
(30):27721–8. https://doi.org/10.1074/jbc.M212065200 PMID: 12761229.
114.
Neff L, Zeisel M, Sibilia J, Scholler-Guinard M, Klein JP, Wachsmann D. NF-kappaB and the MAP
kinases/AP-1 pathways are both involved in interleukin-6 and interleukin-8 expression in fibroblast-like
synoviocytes stimulated by protein I/II, a modulin from oral streptococci. Cellular microbiology. 2001; 3
(10):703–12. PMID: 11580755.
115.
Akira S, Isshiki H, Sugita T, Tanabe O, Kinoshita S, Nishio Y, et al. A nuclear factor for IL-6 expression
(NF-IL6) is a member of a C/EBP family. The EMBO journal. 1990; 9(6):1897–906. PMID: 2112087;
PubMed Central PMCID: PMC551896.
116.
Heinrich PC, Behrmann I, Haan S, Hermanns HM, Muller-Newen G, Schaper F. Principles of interleu-
kin (IL)-6-type cytokine signalling and its regulation. The Biochemical journal. 2003; 374(Pt 1):1–20.
https://doi.org/10.1042/BJ20030407 PMID: 12773095; PubMed Central PMCID: PMC1223585.
117.
Heinrich PC, Behrmann I, Muller-Newen G, Schaper F, Graeve L. Interleukin-6-type cytokine signal-
ling through the gp130/Jak/STAT pathway. The Biochemical journal. 1998; 334 (Pt 2):297–314. PMID:
9716487; PubMed Central PMCID: PMC1219691.
118.
Murakami M, Hibi M, Nakagawa N, Nakagawa T, Yasukawa K, Yamanishi K, et al. IL-6-induced homo-
dimerization of gp130 and associated activation of a tyrosine kinase. Science. 1993; 260(5115):1808–
10. PMID: 8511589.
119.
Harrison DA. The Jak/STAT pathway. Cold Spring Harbor perspectives in biology. 2012; 4(3). https://
doi.org/10.1101/cshperspect.a011205 PMID: 22383755; PubMed Central PMCID: PMC3282412.
120.
Kershaw NJ, Murphy JM, Liau NP, Varghese LN, Laktyushin A, Whitlock EL, et al. SOCS3 binds spe-
cific receptor-JAK complexes to control cytokine signaling by direct kinase inhibition. Nature structural
& molecular biology. 2013; 20(4):469–76. https://doi.org/10.1038/nsmb.2519 PMID: 23454976;
PubMed Central PMCID: PMC3618588.
121.
Blanco-Aparicio C, Renner O, Leal JF, Carnero A. PTEN, more than the AKT pathway. Carcinogene-
sis. 2007; 28(7):1379–86. https://doi.org/10.1093/carcin/bgm052 PMID: 17341655.
122.
Kandel ES, Hay N. The regulation and activities of the multifunctional serine/threonine kinase Akt/
PKB. Experimental cell research. 1999; 253(1):210–29. https://doi.org/10.1006/excr.1999.4690
PMID: 10579924.
123.
Hahn-Windgassen A, Nogueira V, Chen CC, Skeen JE, Sonenberg N, Hay N. Akt activates the mam-
malian target of rapamycin by regulating cellular ATP level and AMPK activity. The Journal of biologi-
cal chemistry. 2005; 280(37):32081–9. https://doi.org/10.1074/jbc.M502876200 PMID: 16027121.
124.
Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3.
Nature reviews Cancer. 2009; 9(11):798–809. https://doi.org/10.1038/nrc2734 PMID: 19851315.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
29 / 30
125.
Giordano V, De Falco G, Chiari R, Quinto I, Pelicci PG, Bartholomew L, et al. Shc mediates IL-6 signal-
ing by interacting with gp130 and Jak2 kinase. Journal of immunology. 1997; 158(9):4097–103. PMID:
9126968.
126.
Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the
treatment of cancer. Oncogene. 2007; 26(22):3291–310. https://doi.org/10.1038/sj.onc.1210422
PMID: 17496923.
127.
Liu H, Yao S, Dann SM, Qin H, Elson CO, Cong Y. ERK differentially regulates Th17- and Treg-cell
development and contributes to the pathogenesis of colitis. European journal of immunology. 2013; 43
(7):1716–26. https://doi.org/10.1002/eji.201242889 PMID: 23620016.
128.
Cruzalegui FH, Cano E, Treisman R. ERK activation induces phosphorylation of Elk-1 at multiple S/T-
P motifs to high stoichiometry. Oncogene. 1999; 18(56):7948–57. https://doi.org/10.1038/sj.onc.
1203362 PMID: 10637505.
129.
Yokogami K, Wakisaka S, Avruch J, Reeves SA. Serine phosphorylation and maximal activation of
STAT3 during CNTF signaling is mediated by the rapamycin target mTOR. Current biology: CB. 2000;
10(1):47–50. PMID: 10660304.
130.
Schuringa JJ, Timmer H, Luttickhuizen D, Vellenga E, Kruijer W. c-Jun and c-Fos cooperate with
STAT3 in IL-6-induced transactivation of the IL-6 respone element (IRE). Cytokine. 2001; 14(2):78–
87. https://doi.org/10.1006/cyto.2001.0856 PMID: 11356008.
A SASP model after DNA damage
PLOS Computational Biology | https://doi.org/10.1371/journal.pcbi.1005741
December 4, 2017
30 / 30
|
29206223
|
Oncogene = ( IL8 ) OR ( IL6 )
p53 = ( ( CHK1 ) AND NOT ( MDM2 ) ) OR ( ( ATM ) AND NOT ( MDM2 ) ) OR ( ( CHK2 ) AND NOT ( MDM2 ) )
ATR = ( DNAD )
CHK1 = ( ATR )
IkB = ( ( NFkB ) AND NOT ( IKK AND ( ( ( NEMO ) ) ) ) ) OR ( ( IkB ) AND NOT ( IKK AND ( ( ( NEMO ) ) ) ) )
p21 = ( p53 ) OR ( HIF1 )
TAK1 = ( TRAF6 ) OR ( TAB )
IKK = ( NEMO ) OR ( NIK ) OR ( Akt )
CHK2 = ( ATM )
MDM2 = ( ( p53 ) AND NOT ( ATM ) )
TRAF6 = ( IRAK )
ATM = ( DNAD )
p38 = ( ( MKK ) AND NOT ( MKP1 ) )
IRAK = ( IRAK ) OR ( MyD88 ) OR ( IL1R )
MyD88 = ( IL1R )
JNK = ( ( MKK ) AND NOT ( MKP1 ) )
Akt = ( PI3K )
RB = NOT ( ( pRB ) OR ( CDK4 ) OR ( CDK2 ) )
MKP1 = ( AP1 )
cJun = ( cFos AND ( ( ( JNK OR CEBPbeta OR p38 OR ERK1_2 ) ) ) )
JAK = ( ( IL6R ) AND NOT ( SOCS3 ) )
STAT3 = ( mTOR ) OR ( cFos AND ( ( ( cJun ) ) ) ) OR ( JAK )
Elk1 = ( ERK1_2 )
NEMO = ( DNAD )
GP130 = ( IL6 )
IL8 = ( CEBPbeta ) OR ( AP1 ) OR ( NFkB )
CDK4 = NOT ( ( p21 ) OR ( p16INK4 ) )
cFos = ( CEBPbeta ) OR ( p38 ) OR ( Elk1 ) OR ( JNK ) OR ( STAT3 )
MEK1_2 = ( GP130 AND ( ( ( IL6 ) ) ) )
IL1R = ( IL1 )
TAB = ( TRAF6 ) OR ( IRAK )
AP1 = ( cJun AND ( ( ( cFos ) ) ) )
p16INK4 = ( Oncogene ) OR ( DNAD )
MEKK = ( TRAF6 )
CDK2 = ( ( E2F ) AND NOT ( p21 ) )
MKK = ( MEKK ) OR ( TAK1 )
mTOR = ( Akt )
IL6 = ( CEBPbeta ) OR ( ERK1_2 ) OR ( NFkB )
IL6R = ( IL6 )
PI3K = ( JAK )
NIK = ( TAK1 )
pRB = ( CDK4 ) OR ( CDK2 )
IL1 = ( NFkB )
SOCS3 = ( STAT3 )
HIF1 = ( ( Hypoxia ) AND NOT ( p53 ) )
ERK1_2 = ( MEK1_2 AND ( ( ( IL6 ) ) ) )
NFkB = ( ( IKK ) AND NOT ( IkB ) )
CEBPbeta = ( Elk1 )
E2F = ( ( E2F ) AND NOT ( RB ) ) OR ( ( pRB ) AND NOT ( RB ) )
|
RESEARCH ARTICLE
A systems pharmacology model for
inflammatory bowel disease
Violeta Balbas-Martinez1,2, Leire Ruiz-Cerda´1,2, Itziar Irurzun-Arana1,2, Ignacio Gonza´lez-
Garcı´a1¤a, An Vermeulen3,4, Jose´ David Go´mez-Mantilla1¤b, Iñaki F. Troco´niz1,2*
1 Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology,
School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain, 2 IdiSNA, Navarra Institute for
Health Research, Pamplona, Spain, 3 Janssen Research and Development, a division of Janssen
Pharmaceutical NV, Beerse, Belgium, 4 Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of
Pharmaceutical Sciences, Ghent, Belgium
¤a Current address: PharmaMar, Colmenar Viejo, Madrid, Spain.
¤b Current address: Boehringer Ingelheim, Ingelheim am Rhein, Germany.
* itroconiz@unav.es
Abstract
Motivation
The literature on complex diseases is abundant but not always quantitative. This is particu-
larly so for Inflammatory Bowel Disease (IBD), where many molecular pathways are qualita-
tively well described but this information cannot be used in traditional quantitative
mathematical models employed in drug development. We propose the elaboration and vali-
dation of a logic network for IBD able to capture the information available in the literature
that will facilitate the identification/validation of therapeutic targets.
Results
In this article, we propose a logic model for Inflammatory Bowel Disease (IBD) which con-
sists of 43 nodes and 298 qualitative interactions. The model presented is able to describe
the pathogenic mechanisms of the disorder and qualitatively describes the characteristic
chronic inflammation. A perturbation analysis performed on the IBD network indicates that
the model is robust. Also, as described in clinical trials, a simulation of anti-TNFα, anti-IL2
and Granulocyte and Monocyte Apheresis showed a decrease in the Metalloproteinases
node (MMPs), which means a decrease in tissue damage. In contrast, as clinical trials have
demonstrated, a simulation of anti-IL17 and anti-IFNγ or IL10 overexpression therapy did
not show any major change in MMPs expression, as corresponds to a failed therapy. The
model proved to be a promising in silico tool for the evaluation of potential therapeutic tar-
gets, the identification of new IBD biomarkers, the integration of IBD polymorphisms to antic-
ipate responders and non-responders and can be reduced and transformed in quantitative
model/s.
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
1 / 19
a1111111111
a1111111111
a1111111111
a1111111111
a1111111111
OPEN ACCESS
Citation: Balbas-Martinez V, Ruiz-Cerda´ L, Irurzun-
Arana I, Gonza´lez-Garcı´a I, Vermeulen A, Go´mez-
Mantilla JD, et al. (2018) A systems pharmacology
model for inflammatory bowel disease. PLoS ONE
13(3): e0192949. https://doi.org/10.1371/journal.
pone.0192949
Editor: Shree Ram Singh, National Cancer Institute,
UNITED STATES
Received: October 17, 2017
Accepted: February 1, 2018
Published: March 7, 2018
Copyright: © 2018 Balbas-Martinez et al. This is an
open access article distributed under the terms of
the Creative Commons Attribution License, which
permits unrestricted use, distribution, and
reproduction in any medium, provided the original
author and source are credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information
files.
Funding: Development of the computational model
was supported by a fellowship grant from the
Navarra Government to Violeta Balba´s-Martı´nez of
61.965 Euros (http://www.navarra.es/home_es/
Actualidad/BON/Boletines/2017/18/Anuncio-5/)
and Janssen Research and Development. The
funders had no role in study design, data collection
and analysis, decision to publish, or preparation of
Introduction
Inflammatory bowel disease (IBD) is a complex gastrointestinal tract disorder characterized by
a functional impairment of the gut wall affecting patients´ quality of life [1,2]. IBD includes
ulcerative colitis (UC) and Crohn’s disease (CD). The natural course of IBD is highly variable
[3–6] and its etiology is still unknown. The incidence of IBD has dramatically increased world-
wide over the past 50 years [7], reaching levels of 24.3 per 100,000 person-years in UC and 20.2
per 100,000 person-years in CD in the developed countries [8].
There is current evidence that Interleukin 6 (IL6), Tumour necrosis factor-alpha (TNFα),
Interferon Gamma (IFNƔ), Interleukin 1 beta (IL1ß), Interleukin 22 (IL22), Interleukin 17
(IL17) and Natural Killer cells (NK), among other signalling pathways, play relevant roles in
the pathogenesis of IBD, which is a reflection of the complexity of that physiological system
[9–12]. That complexity indicates that a universal treatment for IBD may not be feasible for
the vast majority of patients [13,14]. In fact, current biological approved treatments are only
palliative with a high percentage of non-responders. For example, around 50% of IBD patients
treated with the current standard of care, Infliximab (an anti-TNFα) or Vedolizumab (an anti-
α4β7 integrin) do not respond satisfactorily to therapy [15,16]. One characteristic of the cur-
rent IBD biological treatments is that approved therapies target just one signalling pathway,
which might explain the high rate of non-responders and the long-term inefficiency of most
treatments [15,17]. In addition, there is evidence to suggest that optimal treatment for IBD
should involve a combination of different drugs [18,19]. Therefore, there is a need, especially
for complex alterations such as immune-mediated diseases, to change the paradigm of drug
development, considering the main aspects (targets, cross-talking between pathways, therapy
combination) from an integrative and computational perspective.
Given the aforementioned biological complexity of immune-mediated diseases and the fact
that current longitudinal data associated with the most relevant elements of the system are
scarce, a full parameterization of IBD related systems based on a differential equation model
does not yet seem feasible. However, some attempts have been made to describe quantitatively
the IBD systems. For example, Wendelsdorf et al., [20] built a quantitative model based on
ordinary differential equations. However, some key disease elements, such as cytokines and T
cells, were incorporated non-specifically (i.e., all types of cytokine were grouped under the
generic element active cytokines) in the model structure, limiting its use to explore potential
therapeutic targets. More recently, Dwivendi et al., [21], based on the results of a clinical trial
with the anti–IL6R antibody, Tocilizumab, have developed a multiscale systems model in
Crohn’s disease, limited to the IL6–mediated immune regulation pathway.
Network analysis represents a promising alternative in such data limited circumstances
[22–24]. As many molecular pathways in IBD are qualitatively well described, interaction net-
works may be a suitable approach for characterizing IBD. These networks are simplified repre-
sentations of biological systems in which the components of the system such as genes, proteins
or cells are represented by nodes and the interactions between them by edges [25]. Boolean
network models, originally introduced by Kauffman [26,27], represent the simplest discrete
dynamic models. These models only assume two discrete states for the nodes of a network,
ON or OFF, corresponding to the logic values 1 (active) or 0 (not active, but not necessarily
absent) [28]. A well-designed logic model could generate predictive outcomes given a set of
initial conditions. Qualitative, logical frameworks have emerged as relevant approaches with
different applications, as demonstrated by a growing number of published models [29]. Com-
plementing these applications, several groups have provided various methods and tools to sup-
port the definition and analysis of logical models, as it can be seen by the recent achievements
of the Consortium for Logical Models and Tools (CoLoMoTo) in logical modelling [30].
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
2 / 19
the manuscript. Janssen Research and
Development provided support in the form of
salaries for author AV, but did not have any
additional role in the study design, data collection
and analysis, decision to publish, or preparation of
the manuscript. The specific role of this author is
articulated in the ‘author contributions’ section.
Competing interests: We have the following
interests. This study was partly funded by Janssen
Research and Development, the employer of An
Vermeulen. There are no patents, products in
development or marketed products to declare. This
does not alter our adherence to all the PLOS ONE
policies on sharing data and materials, as detailed
online in the guide for authors.
There are already several tools for Boolean modeling of regulatory networks in which it is pos-
sible to define direct activation-inhibition relationships between the components of the net-
work, such as BoolNet R [31] or GINsim [32]. More recently, the R package SPIDDOR
(Systems Pharmacology for effIcient Drug Development On R) among others, has imple-
mented new types of regulatory interactions and perturbations within the system, such as posi-
tive and negative modulators and the polymorphism-like alterations, which lead to richer
dynamics between the nodes [28].
In the specific case of IBD, there have been initial attempts to develop network models. The
multi-state modeling tool published by Mei et al., [33,34] can be considered a proof of concept
in the application of these types of networks in mucosal immune responses. However, the
number of elements that this model considers and integrates is limited for IBD characteriza-
tion, since only six different cytokine types are included in the inter-cellular scale.
The objective of the current manuscript is to present a Boolean based network model incor-
porating the main cellular and protein components known to play a key role in IBD develop-
ment and progression. The model has been built on well-established experimental knowledge,
mostly of human origin, and only including animal data when no other source of information
was available. Our aim has been to build a model structure facilitating key aspects in the treat-
ment of immune mediated disease, such as the selection of the most promising combination
therapies and the study of the impact of polymorphisms on pathway regulation, thus allowing
patient stratification and personalized medicine.
This study provides the scientific community with a (i) computational IBD model imple-
mented in SPIDDOR R package [28], which allows translation of Boolean models (excluding
models enclosing temporal operators) to a standard Markup language in Systems Biology for
qualitative models (SBML qual [35]) which promotes model interoperability, and (ii) a reposi-
tory with the main and updated information known of the immune system and IBD, which
shows model transparency and allows model reusability. The proposed IBD model can be eas-
ily expanded in size and complexity to incorporate new knowledge, or other type of informa-
tion such as proteomic data. The model presented hereafter is general enough to serve as a
skeleton for other relevant immune diseases such as Rheumatoid Arthritis, Psoriasis or Multi-
ple Sclerosis.
The manuscript is organized as follows: In the next section, Results regarding the structure
of the model can be graphically visualized, and the ability of the model to recreate certain alter-
ations that have been reported in IBD is demonstrated, as well as the model’s capability to
reproduce the results from recent clinical trials performed in IBD patients from a high-level
perspective. Applications of the model, including its advantages and limitations are then dis-
cussed together with ideas for future research. Finally, the Methods section provides a detailed
technical description (with the aid of supplementary material) of the network and a descrip-
tion of how simulations, collection, and representation of results have been performed.
Results
Graphical representation, repository, and Boolean functions
The graphical representation of the IBD network is shown in Fig 1. It consists of 43 nodes and
298 qualitative interactions located in three different physiological areas corresponding to (i)
the lymph node, (ii) the blood and lymph circulatory system that irrigates the intestinal epithe-
lial cells and (iii) the gut lumen.
Definition of all nodes and the full documented regulatory interactions conforming the
model structure can be found in supporting information S1 Table and S2 Table, respectively.
The S2 Table is fundamental to understand the rationale for the selection and implementation
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
3 / 19
of the Boolean functions (BF). It was organized to provide a comprehensive summary of the
301 manuscripts (published over the last three decades) used to build the model, highlighting
for example whether (i) a specific pathway was reported to be altered in IBD, or (ii) informa-
tion was supported by human (more than the 80% of the network structure) or animal data.
The Boolean operators used to define the network model of IBD were: the NOT operator
which is noted as “!”, the AND operator which is noted as “&” and the OR operator which is
noted as “|”. Recent and innovative modulators and threshold operators previously described
by Irurzun-Arana et al., 2017 [28] were also part of the arsenal of Boolean elements used in the
model proposed (see S1 File for a detailed description of those additional Boolean elements).
Regarding the input selection, as it is assumed that IBD is caused by intestinal dysbiosis, an
environment of different bacteria was recreated selecting three different antigens which are
components of most Bacterial Gram positive and Gram negative. Therefore, during the devel-
opment of the proposed model the following assumptions were made: First, there is a chronic
exposure to bacterial antigens: Peptidoglycan (PGN), Lipopolysaccharide (LPS) and Muramyl
dipeptide (MDP). PGN is a component of the cell wall of all bacteria, but in particular of
gram-positive bacteria, LPS is a component of the outer membrane of Gram-negative bacteria
Fig 1. Graphical representation of IBD model. Nodes represent cells, proteins, bacterial antigens, receptors or ligands. Bacterial antigens trigger the IBD immune
response through activation of different pattern recognition receptors (TLR2, TLR4 and NOD2) starting the innate and adaptive immune response. Reprinted from [36]
under a CC BY license, with permission from the organizers of the 2016 International Conference on Systems Biology, original copyright 2016.
https://doi.org/10.1371/journal.pone.0192949.g001
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
4 / 19
[37], and MDP is a constituent of both Gram-positive and Gram-negative bacteria [38]. All
three elicit strong immune responses and seem to play a critical role in the development and
pathophysiology of IBD, as it has been hypothesized that the onset or relapse of IBD is trig-
gered by an imbalance in self-microbiota composition than cannot be controlled by immune
system [39]. Table 1 lists the initial conditions expressed by the corresponding BF, and shows
that the nodes representing antigens are chronically expressed unless the natural antimicrobial
peptides perforin (PERFOR), granzyme B (GRANZB) or defensins (DEF) become active.
Second, there is an impairment in antigen elimination in IBD patients [1,40,41], simulated
with the threshold operator Ag_elim = 6. The threshold operator means that PERFOR,
GRANZB, or DEF inhibit antigen activation when any of these three nodes have been activated
for at least 6 consecutive iterations (see Table 1).
Third, the final readout of the network model is the average expression of the output node,
Metalloproteinases (MMPs). There is solid evidence that this group of proteins is directly asso-
ciated with intestinal fibrosis and tissue damage in IBD [42–46] supporting their use as a rele-
vant biomarker in clinical practice as proposed by O’Sullivan et al. [47]. As it can be seen in
Table 2, the nodes that directly activate MMPs are the nodes that have relevant roles in the
pathogenesis of IBD [9–12,42–44,46,48].
Table 2 contains the full set of BF that modulates the signal initialized by the antigens
through the activation of different pattern recognition receptors (TLR2, TLR4 and NOD2
nodes) and the impact on the output node (MMPs) as the recipient of the antigen signal inter-
nal modulation. The nodes TNFα or IFNγ have the most complex pathways as can be seen in
the corresponding Boolean equations (Table 2).
With the aim of making the network model more accessible to the community it has been
uploaded to “The Cell Collective” [49,50] platform (https://www.cellcollective.org/#cb963d7f-
75cb-4b2e-8987-0c7592a9c21d). In addition, the supporting information document S2 File
provides the network model in text format ready for simulation in the R-based freely available
package SPIDDOR [28] and an html tutorial as a guide to reproduce the results (S3 File).
Perturbation analysis and clustering: Network robustness
The results of the network perturbation analysis are presented in Fig 2. The heatmap shows
the impact of a single blockage of each node in every network node. The results indicate that
most node blockages did not trigger considerable changes, suggesting that the IBD network is
robust [51]. Some perturbations led to a higher activation of the nodes, while down regulations
were more common. The heatmap was combined with a hierarchical clustering grouping
together the nodes that caused similar alterations. Knockout of the NFkß node appeared to be
the most relevant alteration as it caused a reduction in expression of many of the nodes that
were reported to be overexpressed in IBD patients. The knockout of the Th0 node (represent-
ing activated CD4+ T cells) also elicited a reduction in MMPs. The positive effects of the NFkß
and Th0 node blockades on MMPs decreased expression, resembled some of the known mech-
anisms of action of glucocorticoids, inhibitors of T cell activation and proinflammatory
Table 1. Boolean functions (BF) of the IBD model to simulate the initial conditions.
INITIAL CONDITIONS: CHRONIC EXPOSURE
PGN ¼ ! ðTAG elim¼6
i¼1
PERFORt i j TAG elim¼6
i¼1
GRANZBt i j TAG elim¼6
i¼1
DEFt iÞ
MDP ¼ ! ðTAG elim¼6
i¼1
PERFORt i j TAG elim¼6
i¼1
GRANZBt i j TAG elim¼6
i¼1
DEFt iÞ
LPS ¼ ! ðTAG elim¼6
i¼1
PERFORt i j TAG elim¼6
i¼1
GRANZBt i j TAG elim¼6
i¼1
DEFt iÞ
https://doi.org/10.1371/journal.pone.0192949.t001
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
5 / 19
Table 2. Boolean functions (BF) of the IBD model for the internal and the output nodes.
INTERNAL NODES
TLR2 = PGN
TLR4 = LPS
NOD2 = MDP
NFkB = TLR2 | NOD2 | TLR4
IL6 = (MACR & PGN) | (DC & (LPS | PGN)) | (Th17 & IL23) | (NFkB &! (IL4 | IL10))
TNFa ¼ ððNFkB&LPSÞ j ðMACR&ðIL2 j ðIFNg&LPSÞ j PGNÞÞ j ðNK&ðMDP j PGN j LPSÞ&ððIL2 j
IL12Þ&ðIL2 jIL15ÞÞ j ðFIBROBLAST&IFNgÞ j ððCD4 NKG2D j CD8 NKG2D j NK NKG2DÞ&ðIEC MICA B j
IEC ULPB1 6ÞÞÞ&! ðTdownregcyt¼4
i¼1
IL10t i&ðTdownregcyt¼4
i¼1
TLR2t ij Tdownregcyt¼4
i¼1
TLR4t iÞ&TNFaÞ
TGFb = (Treg | MACR)
Th0 ¼ TTHR
Th0¼3
i¼1
LPSt ij TTHR
Th0¼3
i¼1
MDPt ij TTHR
Th0¼3
i¼1
PGNt i
Th0_M = (Th0 & (IL23 | IL12)) | Th0_M
IL18 = ((MACR | DC) & LPS) & NFkB
IL1b ¼ ððMACR j DCÞ&LPS&NFkBÞ&! ðIL1b&Sdownreg
cyt¼4
i¼1
IL10t iÞ
IFNg ¼ ððNK&ðPGNjLPSjMDPj&ðIL23jðIL12&ðIL2jIL15 jIL18ÞÞÞÞ j ðTh0 M&ðLPS j MDP j PGNÞ
&ðIL12 j IL23ÞÞ j Th1 j ððCD8 NKG2D j NK NKG2DÞ&ðIEC MICA B j IEC ULPB1 6ÞÞ j ðTh17&ðPGN j LPSj
MDPÞÞ j ððMACR j Th0Þ&IL18&IL12ÞÞ&! ððIFNg&ðTdownreg
cyt¼4
i¼1
TGFbt i j Tdownregcyt¼4
i¼1
IL10t i j Th2Þ
IL23 = (MACR & IL1b) | DC
IL22 ¼ Th17jðNK&ððIL18&IL12Þ j IL23ÞÞjCD4 NKG2DjðððIL22&Th0&IL21Þ&!ðTupreg
cyt¼3
i¼1
IL22t i&
Tupreg
cyt¼3
i¼1
Th0t i&Tupreg
cyt¼3
i¼1
IL21t iÞÞ&! TGFbÞ
IL21 = Th17 | ((Th0 & IL6) &! (IL4 | IFNg | TGFb))
IL17 ¼ ðTh17 j ðTh17 M&ðLPS j MDP j PGNÞÞ j ðCD4 NKG2D&ðIEC MICA B j IEC ULPB1 6ÞÞÞ&!
ððTdownreg
cyt¼4
i¼1
TGFbt i j Tdownreg
cyt¼4
i¼1
IL13t iÞ&IL17Þ
IL10 = Treg|(Th2 &! IL23)|((TLR2 & NFkB) &! (MACR & IFNg)) | ((MACR & LPS) &! IL4) | (DC & LPS)
Th17 ¼ ððTh0&ðIL1b j IL23 j IL6ÞÞ j ððTh17&IL23Þ&!ðTupreg
cell¼2
i¼1
Th17t i&Tupreg
cell¼2
i¼1
Il23t iÞÞÞ&!
ððSdownreg
cell¼2
i¼1
TGFbt ij Sdownreg
cell¼2
i¼1
IL12t i j Sdownreg
cell¼2
i¼1
IL4t ij Sdownreg
cell¼2
i¼1
IFNgt i j
Sdownreg
cell¼2
i¼1
Treg t iÞ&Th17Þ
Th17_M = ((Th0_M & (PGN | MDP | LPS)) & ((IL1b & IL6) | IL23 | IL2)) | Th17_M
Th1 ¼ ðTh0&ððIL12 j IFNg j IL18Þ j ðDC&IL12&IL23&LPSÞÞÞ&! ðððSdownreg
cell¼2
i¼1
IL17t i&
Sdownreg
cell¼2
i¼1
IL12t iÞ j ðSdownreg
cell¼2
i¼1
Treg t ij Sdownreg
cell¼2
i¼1
Th2t ij Sdownreg
cell¼2
i¼1
TGFbt i j
Sdownreg
cell¼2
i¼1
IL10t ij Sdownreg
cell¼2
i¼1
IL4t iÞÞ&Th1Þ
Th2 ¼ ðTh0&ðIL10 jððIL18&IL4Þ&!IL12ÞÞj ððTh2&IL4Þ&!ðTupreg
cell¼2
i¼1
Th2t i&Tupreg
cell¼2
i¼1
IL4t iÞÞÞ&!
ððSdownreg
cell¼2
i¼1
Tregt ij Sdownreg
cell¼2
i¼1
IFNgt ij Sdownreg
cell¼2
i¼1
TGFbt iÞ&Th2Þ
IL4 = Th2
IL15 = (FIBROBLAST & (MDP | LPS | PGN)) | (MACR & (LPS | IFNg))
IL12 ¼ ððððMACR j DCÞ&ðLPS jPGNÞ&IFNgÞ&!ðIL12&Sdownreg
cyt¼4
i¼1
TNFat iÞÞ j ðDC&IL1bÞ j
ðIL12&ðIL13 j IL4ÞÞÞ&!ððSdownreg
cyt¼4
i¼1
TGFbt i j Sdownreg
cyt¼4
i¼1
IL10t iÞ&IL12Þ
IL13 = Th2
Treg ¼ ðTTHR
Th0
Treg¼3
i¼1
Th0t i&ðTGFb j TLR2ÞÞ&! ððSdownreg
cell¼2
i¼1
IL6t ij Sdownreg
cell¼2
i¼1
IL21t ij
Sdownreg
cell ¼2
i¼1
IL23t ij Sdownreg
cell¼2
i¼1
Th17t ij Sdownreg
cell¼2
i¼1
IL22t ij Sdownreg
cell¼2
i¼1
TNFat iÞ&TregÞ
NK ¼ ðIL15 jIL2 j IL12 jIL23j ðIL18&IL10ÞÞ&! ðSdownregcell¼2
i¼1
Tregt i&NKÞ
DEF ¼ IL22 j IL17 j TTHR
NOD2
DEF¼3
i¼1
NOD2t i
IL2 = Th0 | (Th0_M & (MDP | LPS | PGN)) | DC
MACR ¼ ðNFkB j ððMACR&ðIFNg jIL15ÞÞ&! ðTupreg
cell¼2
i¼1
NFkBt i&ðTupreg
cell¼2
i¼1
IFNgt i j
Tupreg
cell¼2
i¼1
IL15t iÞÞÞÞ&! ðSdownreg
cell¼2
i¼1
IL10t i&MACRÞ
DC ¼ NFkB&! ðSdownreg
cell¼2
i¼1
IL10t i&DCÞ
IEC MICA B ¼ ððLPS j MDP j PGNÞ j ðIEC MICA B&TNFaÞ&! ðTupreg
rec¼2
i¼1
IEC MICA Bt i&
Tupreg
rec¼2
i¼1
TNFat iÞÞ&! TGFb
IEC_ULPB1_6 = CD8_NKG2D & (LPS|MDP|PGN)
CD8 NKG2D ¼ ðLPS j PGN j MDPÞ&!ððTTHR
LIGANDS
NKG2D¼3
i¼1
IEC MICA Bt ij TTHR
LIGANDS
NKG2D¼3
i¼1
IEC ULPB 1 6t i
j ðSdownreg
cell¼2
i¼1
IL21t i&Sdownreg
cell¼2
i¼1
IL2t iÞÞ&CD8 NKG2DÞ
NK NKG2D ¼ ðLPSjPGNjMDPÞ&! ðSdownreg
cell¼2
i¼1
TGFbt ij TTHR
LIGANDS
NKG2D¼3
i¼1
IEC MICA Bt i j
TTHR
LIGANDS
NKG2D¼3
i¼1
IEC ULPB 1 6t i jðSdownreg
cell¼2
i¼1
IL21t i&Sdownreg
cell¼2
i¼1
IL12t iÞÞ&NK NKG2DÞ
CD4 NKG2D ¼ ðLPS j PGN j MDP j ðCD4 NKG2D&ðIL15 j TNFaÞÞ&! ðTupreg
rec¼2
i¼1
CD4 NKG2Dt i
&ðTupreg
rec¼2
i¼1
IL15t ij Tupreg
rec¼2
i¼1
TNFat iÞÞÞ&! ððSdownreg
cell¼2
i¼1
IL10t i j
TTHR
LIGANDS
NKG2D¼3
i¼1
IEC MICA Bt i j TTHR
LIGANDS
NKG2D¼3
i¼1
IEC ULPB 1 6t iÞ&CD4 NKG2DÞÞ
FIBROBLAST ¼ ððMACR&ðIL4 j IL13 j TGFbÞÞjIL2Þ&!ððSdownreg
cell¼2
i¼1
IFNgt ij Sdownreg
cell¼2
i¼1
IL12t iÞ
&FIBROBLASTÞ
PERFOR = NK | NK_NKG2D
(Continued)
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
6 / 19
cytokines, as well as potent suppressors of the effector function of monocyte-macrophage and
fibroblastic activity, interfering with the NFκB inflammatory signal [52–54].
Network accuracy and validation
Experimental and clinical information.
Simulations of chronic infection in IBD individ-
uals show that the model reproduced satisfactorily experimental and clinical information
(summarized in Table 3 and supporting information S3 Table). Fig 3 shows the results of the
simulation for each network node after reaching the attractor state for virtual healthy and IBD
subjects. In total, 31 upregulations in experimental studies were replicated with our simula-
tions. Similarly, the 9 nodes reported as altered appeared upregulated in the simulations, and
finally, the three nodes whose profiles were not known also proved to be upregulated.
Clinical trials.
In our simulations, three drugs that have failed to prove clinical efficacy in
clinical trials (anti-IL17, anti-IFNγ and rhuIL-10) also exhibited no benefit in the simulated
surrogate for the disease score (Fig 4). Simulations with anti-TNFα, a biologic therapy
approved for IBD, showed a decrease in the disease score. Simulations with anti-IL12-IL23, a
recently approved therapy for IBD, showed a slight decrease in MMPs and anti-IL2 therapy
simulation showed a decrease similar to anti-TNFα. In addition, the new promising therapy
(GMA), equivalent to an anti-MACR in our model showed a decrease in MMPs similar to that
for anti-TNFα.
Discussion
In the current study, we present a Systems Pharmacology (SP) network model for IBD based
on the main cells and proteins involved in the disease. Our analysis appears timely, as IBD has
recently been attracting increasing attention [55–59]. We attempted to meet one of the major
challenges in inflammatory bowel disease (IBD) which is the integration of IBD-related infor-
mation to construct a predictive model. We are not the only ones following this line of
research, as Lauren A Peters et al. have very recently performed a key driver analysis to identify
the genes predicted to modulate network regulatory states associated with IBD [55]. Both anal-
yses could be integrated in the future and inform our post-transcriptomic network with the
key driver genes identified by Lauren A Peters et al. [55].
In comparison with the previous quantitative approaches for IBD [20,21,33,34], our model
identified Naive CD4+ T Cells, Macrophages and Fibroblasts cells as relevant in IBD. Also, in
addition to the six interleukins (TGFß, IL6, IL17, IL10, IL12 and IFNγ) considered by Mei
et al. [33,34] our network involves 10 interleukins more which could represent possible IBD
biomarkers [60]. The procedure to evaluate the potential role of the different components on
the disease as plausible biomarkers, would be equal to the one described in section 4.5 (pertur-
bation analysis and clustering), focussing on the changes in the output node.
In the validation of network models, robustness and practical applicability represent critical
aspects. The fact that the information gathered from the literature was obtained under very
Table 2. (Continued)
GRANZB = CD8_NKG2D | NK | NK_NKG2D | (DC &! (LPS | PGN))
OUTPUT NODE
MMPs = (MACR & TNFa) | (FIBROBLAST & (IL21 | IL17 | IL1b | TNFa))
Bold text within Boolean equations indicates that the information belongs to animal data
https://doi.org/10.1371/journal.pone.0192949.t002
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
7 / 19
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
8 / 19
different experimental designs/conditions/methodologies, represents a challenge with respect
to validation. This led us to propose and adopt a novel strategy consisting of the comparison of
the results of model-based virtual pathway simulations with those reported in the literature for
IBD patients. Using this approach, we obtained a qualitative reproduction of IBD in which all
the network elements that have been reported as upregulated in IBD patients appeared upre-
gulated in our simulation results. The perturbation analysis of the network was performed by a
single blockage in each node to analyse how that type of alteration propagates through the
entire network reflecting the case of single polymorphisms, which represents the simplest case
of IBD disease. Despite of the simplicity of this analysis, the results obtained from the model
accuracy and validation procedures are encouraging. Results from the perturbation analysis
indicate that the proposed network model is robust, as alteration in most nodes did not trigger
considerable changes in the network [61].
Once validated and checked for robustness, the network was challenged to qualitatively
reproduce the readouts of five different therapies reported in experimental and clinical studies.
The outcome of this challenge was similar to the clinical output in IBD patients. By the simula-
tion of TNFα or MACR knockout (simulating Granulocyte and Monocyte Apheresis), a
decrease in MMPs node was observed, which is in line with therapy success in clinical practice
by a decrease in Crohn’s Disease Activity Index (CDAI) Score [42–46],[62–68]. On other
hand, IL17 or IFNγ knockout or IL10 overexpression did not show major change in MMPs
expression, suggested a failed therapy as was indeed found in clinical practice [69–72].
Surprisingly, the model shows that a knockout of IL2 leads to a reduction in MMPs similar
to that of a knockout of TNFα, even when previous results of clinical trials with Basiliximab or
Daclizumab (monoclonal antibodies that bind to the interleukin 2 receptor CD25) in Ulcera-
tive Colitis have failed to show superiority to corticosteroids alone [73,74]. The mechanism of
Fig 2. IBD network perturbation analysis and clustering. The heatmap indicates the effect of single blockage of each node (columns) in every network
node (rows). The colour in each cell corresponds to the Perturbation Index (PI) of the nodes. When there is no change in the expression of the node, the
cells of the heatmap would be black, having a value between 0.8 and 1.25 in their PIs. Otherwise, when the perturbation causes an overexpression in a
node, the cell in the heatmap would be orange coloured, with PIs values greater than 1.25. On the contrary, a value of 0.8 or smaller, blue colour, indicates
that the perturbation causes a downregulation of the node. The numeric scale in the legend represents different values of the nodes PI under different
perturbations. Nodes that induce similar alterations are hierarchically clustered.
https://doi.org/10.1371/journal.pone.0192949.g002
Table 3. Expression of network nodes in IBD patients.
NODE
EXPRESSION
NODE
EXPRESSION
NODE
EXPRESSION
NODE
EXPRESSION
PGN
MDP
LPS
Altered
IL1b
Upregulated
Th2
Upregulated
DC
Downregulated in Blood-Upregulated in
mucosa
TLR2
Upregulated
IFNg
Upregulated
IL4
Altered
IEC_MICA_B
Upregulated
TLR4
Upregulated
IL23
Upregulated
IL15
Upregulated
IEC_ULPB1_6
Upregulated
NOD2
Altered
IL22
Upregulated
IL12
Upregulated
CD8_NKG2D
Upregulated
NFkB
Altered
IL21
Upregulated
IL13
Upregulated
NK_NKG2D
Unknown
IL6
TNFa
Upregulated
Upregulated
IL17
Upregulated
Treg
Downregulated in Blood-Upregulated in
mucosa
CD4_NKG2D
Upregulated
TGFb
Upregulated
IL10
Upregulated
NK
Upregulated
FIBROBLAST
Upregulated
Th0
Unknown
Th17
Upregulated
DEF
Altered
MMPs
Upregulated
Th0_M
Upregulated
Th17_M
Upregulated
IL2
Upregulated
PERFOR
Altered
IL18
Upregulated
Th1
Altered
MACR
Unknown
GRANZB
Upregulated
A total of 31 nodes are reported as upregulated in IBD patients, 9 are reported to be altered (when different reports from literature are inconclusive or contradictory)
and 3 nodes are unknown.
https://doi.org/10.1371/journal.pone.0192949.t003
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
9 / 19
action of corticosteroids has not been fully described, yet it is known that corticosteroids cause
diminished levels of IL2 mRNA [75,76]. Together with the rest of corticosteroid inhibitory
mechanisms, this would be the reason why Basiliximab or Daclizumab do not show superiority
to corticosteroids alone.
Among the potential applications the current network supports: (i) biomarker selection
given that the cytokines TNFα, IL21, IL17 and IL1ß, which can be easily measured in periph-
eral plasma with different Enzyme-linked immunosorbent assay (ELISA) kits [77,78], are the
model components directly related to MMPs activation, (ii) search for optimal combination
therapy to overcome the high attrition rates in phase clinical trials with single therapies which
Fig 3. IBD network simulation results. Attractor state of every network node for healthy and IBD simulated individuals under chronic antigen exposure.
https://doi.org/10.1371/journal.pone.0192949.g003
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
10 / 19
are due mainly to lack of efficacy [79], and (iii) management of multiscale information such as
the integration of proteomic gene expression data [55] accounting for IBD polymorphisms to
anticipate responders and non-responders. With such a type of data able to correlate a genetic
alteration with a decrease or an increase in protein expression, it would be possible to simulate
specific genetic alteration by altering the protein expression. This would allow one of the limi-
tations of the current network at the present time to be overcome with regard to the effects of
Ustekinumab, a monoclonal antibody targeting free IL12 and IL23, which has been recently
approved for moderately to severely active Crohn’s disease in adults who have failed to treat-
ment with immunomodulators, or more than one TNFα blocker [80]. Simulation results
based on the known mechanisms of Ustekinumab showed just a 4.1% decrease in tissue dam-
age. On the other hand, when simulating TNFα blocker effects, tissue damage decreased by
30.6% even though a substantial percentage of patients showed poor control of the disease
after treatment with anti-TNFα antibody [15,16].
We emphasize that the proposed network model is fully accessible which allows it to
undergo immediate testing and further development. In that respect it should be noted that
although our model intended to include information of human origin exclusively, some criti-
cal pathways had to be complemented with animal derived data (although in the current case
the percentage of human supported pathways is greater than in previous computational mod-
els [20,81,82]), but we are aware of the wide differences in the immune system between species
[83–85].
Fig 4. Comparison of MMPs expression after the simulation in IBD simulated individuals of different therapies.
Simulated therapies: Anti-TNFα, GMA therapy (equivalent of knock out our MACR node), anti-IL17, human
recombinant IL10 (rhulL-10), anti-IFNγ, anti-IL2 and anti-IL12-IL23. Comparing with untreated simulation, we can
see a 30.7%, a 27.1%, a 31.9% and a 4.1% decrease in the MMPs expression simulating anti-TNFα, GMA therapy, anti-
IL2 and anti-IL12-IL23 respectively. There is no major change in MMPs expression for the two which failed in clinical
trials anti-IL17 (a 6.5% decrease) and human recombinant IL10 (a 3.2% decrease). Otherwise, anti-IFNγ therapy
simulation shows an increase in MMPs expression of 16.0% compared to Untreated.
https://doi.org/10.1371/journal.pone.0192949.g004
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
11 / 19
This study addresses the goals of systems pharmacology by effectively encompassing prior
knowledge to generate a mechanistic and predictive understanding at the systems level for
IBD. Semi-quantitative understanding at the network level is necessary prior to the generation
of detailed quantitative models for within-host disease dynamics. The current IBD model and
the companion literature summary archive will drive the development of a dynamic (i.e., ordi-
nary differential equation driven) model involving meaningful parameters capable of simulat-
ing longitudinal data, and allowing model reduction as well the goal of parameter estimation
during the clinical stages of the drug development process. In addition, our IBD network can
be extended to other inflammatory diseases, as main pathways in the model are common to
most inflammatory conditions [86,87], and the outputs of our nodes could also serve as inputs
to broader-scale logic models; for example, incorporating structures from available logic mod-
els of some of our nodes such as fibroblast [61], IL1b or IL6 [88].
In summary, we present a network model for inflammatory bowel disease which is available
and ready to be used and can cope with (multi-scale) model extensions. It is supported by a
comprehensive repository summarizing the results of the most relevant literature in the field.
This model proved to be promising for the in silico evaluation of potential therapeutic targets,
the search for pathway specific biomarkers, the integration of polymorphisms for patient strat-
ification, and can be reduced and transformed in quantitative model/s.
Methods
Literature search and data selection
The network model is based on an exhaustive bibliographic review focusing on the essential
components of IBD, as previously performed by Ruiz-Cerda´ et al., in their systems pharmacol-
ogy approach for lupus erythematosus [23]. Our review included around 620 papers published
between October 1984 and September 2017, yet the most common reviewed articles were from
2007 or later (76%). The search of the relevant literature was made through Medical Subject
Headings (MeSH) terms using different search engines such as PubMed, clinicaltrials.gov or
google scholar. MeSH terms were focused on the combination of keywords and free words
including: (i) relevant network components (ej.”IL6”) involved in the pathogenesis of IBD, (ii)
nodes that have been reported to be altered in IBD (ej. “IL6 AND IBD”) and (iii) nodes directly
affecting the expression of the nodes selected in (i) and (ii) (ej. “DC AND IL6”). The internal
nodes selection was made according to the reported upregulated components in IBD patients
together with the nodes (immune system cells) which are necessary to link the upregulated
nodes, which were established as internal nodes. Only original papers with a clear description
of experimental conditions were considered to identify the relationships between the compo-
nents of the biological network. Due to the reported differences between animal and human
immunology [83–85], in only few cases were animal data considered to connect nodes of criti-
cal pathways when no human data were available.
Annotation and system representation
Annotation was crucial to organize the available literature according to its relevance. S2 Table
from supplementary information shows the way the information was organized for building
the network. S2 Table includes every node definition and the relationships between the nodes.
Annotation included the identification of the main elements (antigens, cytokines, cells, pro-
teins, membrane receptors and ligands) of IBD disease.
The IBD model will be freely accessible to the public through the “The Cell Collective”
repository https://cellcollective.org/#cb963d7f-75cb-4b2e-8987-0c7592a9c21d.
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
12 / 19
Boolean network building and r implementation
The collection of qualitative relationships extracted from the literature was transformed into a
logical model as described before by Ruiz-Cerda´ et al. [23]. Logic networks capture the dynam-
ics of their components, called nodes, after selected stimuli or initial conditions [89,90]https://
paperpile.com/c/XvtklO/p0BRz+YiQ4q. In these models the relationships of activation or
inhibition between nodes are described as combinations of the logic operators: AND, OR and
NOT condensed in a mathematical expression called a Boolean function for each node. Posi-
tive and negative modulators, and thresholds as previously described by Ruiz-Cerda´ et al.[23]
and Irurzun-Arana et al. [28] were also considered to resemble better the biological system.
Boolean network building and R implementation from S1 File gives a more detailed explana-
tion of the modulators used in the model.
Simulations
The set of combined Boolean functions for the IBD model was implemented SPIDDOR [28],
using RStudio Version 0.99.442. Simulations with 25 repetitions over 5000 iterations were per-
formed. According to preliminary experiments, these simulation conditions were required to
achieve the steady state of the network called attractor [91–93]. An attractor can be a fixed-
point if it composed of one state, a simple cycle if consists of more than one state that oscillates
in a cycle or a complex attractor if a set of steady-states oscillate irregularly. In each simulation,
a node can show two possible values in each iteration: 0 (deactivated) or 1 (activated). The per-
centage of activation of the output node (MMPs) calculated at the attractor state was used as
the readout summary of the simulation exercises, as this group of proteins are directly associ-
ated with intestinal fibrosis and tissue damage in IBD [42–46].
Each node was updated asynchronously [94–96] according to its Boolean function that
defines the dynamics of the system. Initial conditions are explained in detail in “Simulations”
from S1 File.
Perturbation analysis and clustering
Robustness can be defined as the system’s ability to function normally under stochastic pertur-
bations [96]. The investigation of robustness in Boolean networks generally focuses on the
dependence between robustness and network connectivity [97]. We performed a perturbation
analysis in our IBD model to study robustness by simulating the effect of the single blockage of
each node on every other node of the network [51]. This simulation was performed by using
the KO_matrix.f function from SPIDDOR package with 24 repetitions over 999 iterations
under asynchronous updating.
Results from the simulations described above were represented as heatmaps with dendro-
grams in which the number of rows and columns is equal to the number of nodes in the net-
work (Fig 2). The colour in each cell of the heatmap corresponds to the Perturbation Index(PI)
of the nodes, which is the probability ratio between the perturbed and the normal conditions
as described by Irurzun-Arana et al. [28]. A hierarchical clustering method [98] was applied to
further study which nodes cause similar alterations in the system.
Network accuracy and validation
Accuracy was evaluated comparing the alterations reported in the literature for IBD patients
with the simulations of chronic antigen exposure for IBD or healthy individuals.
A literature search of every node expression in IBD patients was performed, and the gath-
ered information is condensed in S3 Table including three categories: up-, down-regulated, or
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
13 / 19
altered, whether the levels in CD, UC or both (IBD) with respect to healthy volunteers are
higher, lower, or inconclusive and/or contradictory, respectively.
For validation purposes, model simulations were compared against available results from
clinical trials performed in IBD, CD or UC until the beginning of 2017 in https://www.
clinicaltrials.gov/. All the molecules tested in clinical trials, whose mechanism of action is
known and whose target were included in our network, were tested with the model. The net-
work was evaluated comparing simulations and reported outcomes from clinical trials for six
investigated molecules: anti-TNFα [62–65] and anti-IL12-IL23 [80], two monoclonal antibod-
ies (mAb) approved for IBD disease, anti-IFNγ [69,70], anti-IL17 [72], anti-IL2 [73,74] and
human recombinant IL10 (rhuIL-10) [71] which failed in clinical trials. Also a new promising
therapy: Granulocyte and Monocyte Apheresis (GMA) [66–68] was tested. The reported
CDAI (Crohn Disease Activity Index) was compared with the average expression of the MMPs
output node in the attractor state.
Supporting information
S1 Table. Abbreviations. List of abbreviations.
(PDF)
S2 Table. IBD Network Repository. Table of nodes and interactions supported by references.
(PDF)
S3 Table. IBD_validation. Table of alterations in patients of IBD network nodes supported by
references.
(PDF)
S1 File. Supporting_Information_Methods. Document with detailed description of the
methodology.
(DOCX)
S2 File. IBD.txt. Text document with the Boolean functions written in SPIDDOR nomencla-
ture for iBD simulation.
(TXT)
S3 File. User_Guide_SPIDDOR_IBD.html. Html tutorial about how to reproduce the results
from the present manuscript with the SPIDDOR package.
(HTML)
Acknowledgments
We would like to thank The Cell Collective team, specially to Tomas Helikar, for their help in
building the model in their platform and making it more accessible to the community.
Author Contributions
Conceptualization: Violeta Balbas-Martinez, Jose´ David Go´mez-Mantilla, Iñaki F. Troco´niz.
Data curation: Violeta Balbas-Martinez, Leire Ruiz-Cerda´, Ignacio Gonza´lez-Garcı´a.
Formal analysis: Violeta Balbas-Martinez, Leire Ruiz-Cerda´, Itziar Irurzun-Arana, Jose´ David
Go´mez-Mantilla.
Funding acquisition: An Vermeulen, Iñaki F. Troco´niz.
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
14 / 19
Investigation: Violeta Balbas-Martinez, Leire Ruiz-Cerda´, Ignacio Gonza´lez-Garcı´a, Jose´
David Go´mez-Mantilla.
Methodology: Violeta Balbas-Martinez, Itziar Irurzun-Arana, Ignacio Gonza´lez-Garcı´a, Jose´
David Go´mez-Mantilla, Iñaki F. Troco´niz.
Project administration: An Vermeulen, Iñaki F. Troco´niz.
Software: Violeta Balbas-Martinez, Itziar Irurzun-Arana, Ignacio Gonza´lez-Garcı´a, Jose´ David
Go´mez-Mantilla.
Supervision: An Vermeulen, Jose´ David Go´mez-Mantilla, Iñaki F. Troco´niz.
Validation: Violeta Balbas-Martinez, Jose´ David Go´mez-Mantilla.
Visualization: Violeta Balbas-Martinez.
Writing – original draft: Violeta Balbas-Martinez, Iñaki F. Troco´niz.
Writing – review & editing: Violeta Balbas-Martinez, Jose´ David Go´mez-Mantilla, Iñaki F.
Troco´niz.
References
1.
Wehkamp J, Go¨tz M, Herrlinger K, Steurer W, Stange EF. Inflammatory Bowel Disease. Dtsch Arztebl
Int. 2016; 113: 72–82. https://doi.org/10.3238/arztebl.2016.0072 PMID: 26900160
2.
Matricon J, Barnich N, Ardid D. Immunopathogenesis of inflammatory bowel disease. Self Nonself.
2010; 1: 299–309. https://doi.org/10.4161/self.1.4.13560 PMID: 21487504
3.
Solberg IC, Lygren I, Jahnsen J, Aadland E, Høie O, Cvancarova M, et al. Clinical course during the first
10 years of ulcerative colitis: results from a population-based inception cohort (IBSEN Study). Scand J
Gastroenterol. 2009; 44: 431–440. https://doi.org/10.1080/00365520802600961 PMID: 19101844
4.
Faubion WA Jr, Loftus EV Jr, Harmsen WS, Zinsmeister AR, Sandborn WJ. The natural history of corti-
costeroid therapy for inflammatory bowel disease: a population-based study. Gastroenterology. 2001;
121: 255–260. PMID: 11487534
5.
Loftus EV, Schoenfeld P, Sandborn WJ. The epidemiology and natural history of Crohn’s disease in
population-based patient cohorts from North America: a systematic review. Aliment Pharmacol Ther.
Wiley Online Library; 2002; 16: 51–60. PMID: 11856078
6.
Henriksen M, Jahnsen J, Lygren I, Sauar J, Kjellevold Ø, Schulz T, et al. Ulcerative colitis and clinical
course: results of a 5-year population-based follow-up study (the IBSEN study). Inflamm Bowel Dis.
2006; 12: 543–550. https://doi.org/10.1097/01.MIB.0000225339.91484.fc PMID: 16804390
7.
Gasparetto M, Guariso G. Highlights in IBD Epidemiology and Its Natural History in the Paediatric Age.
Gastroenterol Res Pract. 2013; 2013: 829040. https://doi.org/10.1155/2013/829040 PMID: 24454343
8.
Molodecky NA, Soon S, Rabi DM, Ghali WA, Ferris M. Increasing incidence and prevalence of the
inflammatory bowel diseases with time, based on systematic review. Gastroenterology. Elsevier; 2012;
Available: http://www.sciencedirect.com/science/article/pii/S0016508511013783
9.
Brand S, Beigel F, Olszak T, Zitzmann K, Eichhorst ST, Otte J-M, et al. IL-22 is increased in active
Crohn’s disease and promotes proinflammatory gene expression and intestinal epithelial cell migration.
Am J Physiol Gastrointest Liver Physiol. 2006; 290: G827–38. https://doi.org/10.1152/ajpgi.00513.2005
PMID: 16537974
10.
Yadav PK, Chen C, Liu Z. Potential role of NK cells in the pathogenesis of inflammatory bowel disease.
J Biomed Biotechnol. 2011; 2011: 348530. https://doi.org/10.1155/2011/348530 PMID: 21687547
11.
Rovedatti L, Kudo T, Biancheri P, Sarra M, Knowles CH, Rampton DS, et al. Differential regulation of
interleukin 17 and interferon gamma production in inflammatory bowel disease. Gut. 2009; 58: 1629–
1636. https://doi.org/10.1136/gut.2009.182170 PMID: 19740775
12.
Sartor RB. Mechanisms of disease: pathogenesis of Crohn’s disease and ulcerative colitis. Nat Clin
Pract Gastroenterol Hepatol. 2006; 3: 390–407. https://doi.org/10.1038/ncpgasthep0528 PMID:
16819502
13.
Hamedani R, Feldman RD, Feagan BG. Review article: Drug development in inflammatory bowel dis-
ease: budesonide—a model of targeted therapy. Aliment Pharmacol Ther. 1997; 11 Suppl 3: 98–107;
discussion 107–8.
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
15 / 19
14.
de Lange KM, Barrett JC. Understanding inflammatory bowel disease via immunogenetics. J Autoim-
mun. 2015; https://doi.org/10.1016/j.jaut.2015.07.013 PMID: 26257098
15.
Naija N, Karoui S, Serghini M, Kallel L, Boubaker J, Filali A. [Management of failure of infliximab in
inflammatory bowel disease]. Tunis Med. 2011; 89: 517–521. PMID: 21681712
16.
Shelton E, Allegretti JR, Stevens B, Lucci M, Khalili H, Nguyen DD, et al. Efficacy of Vedolizumab as
Induction Therapy in Refractory IBD Patients: A Multicenter Cohort. Inflamm Bowel Dis. 2015; 21:
2879–2885. https://doi.org/10.1097/MIB.0000000000000561 PMID: 26288002
17.
Raine T. Vedolizumab for inflammatory bowel disease: Changing the game, or more of the same?
United European Gastroenterol J. 2014; 2: 333–344. https://doi.org/10.1177/2050640614550672
PMID: 25360311
18.
Coskun M, Vermeire S, Nielsen OH. Novel Targeted Therapies for Inflammatory Bowel Disease.
Trends Pharmacol Sci. 2016; https://doi.org/10.1016/j.tips.2016.10.014 PMID: 27916280
19.
Grevenitis P, Thomas A, Lodhia N. Medical Therapy for Inflammatory Bowel Disease. Surg Clin North
Am. 2015; 95: 1159–82, vi. https://doi.org/10.1016/j.suc.2015.08.004 PMID: 26596920
20.
Wendelsdorf K, Bassaganya-Riera J, Hontecillas R, Eubank S. Model of colonic inflammation: immune
modulatory mechanisms in inflammatory bowel disease. J Theor Biol. 2010; 264: 1225–1239. https://
doi.org/10.1016/j.jtbi.2010.03.027 PMID: 20362587
21.
Dwivedi G, Fitz L, Hegen M, Martin SW, Harrold J, Heatherington A, et al. A Multiscale Model of Inter-
leukin-6–Mediated Immune Regulation in Crohn’s Disease and Its Application in Drug Discovery and
Development. CPT: Pharmacometrics & Systems Pharmacology. 2014; 3: 1–9.
22.
Chen S, Jiang H, Cao Y, Wang Y, Hu Z, Zhu Z, et al. Drug target identification using network analysis:
Taking active components in Sini decoction as an example. Sci Rep. 2016; 6: 24245. https://doi.org/10.
1038/srep24245 PMID: 27095146
23.
Ruiz-Cerda´ ML, Irurzun-Arana I, Gonza´lez-Garcia I, Hu C, Zhou H, Vermeulen A, et al. Towards patient
stratification and treatment in the autoimmune disease lupus erythematosus using a systems pharma-
cology approach. Eur J Pharm Sci. 2016; https://doi.org/10.1016/j.ejps.2016.04.010 PMID: 27080094
24.
Zhou W, Wang Y, Lu A, Zhang G. Systems Pharmacology in Small Molecular Drug Discovery. Int J Mol
Sci. 2016; 17: 246. https://doi.org/10.3390/ijms17020246 PMID: 26901192
25.
Le Novère N. Quantitative and logic modelling of molecular and gene networks. Nat Rev Genet. 2015;
16: 146–158. https://doi.org/10.1038/nrg3885 PMID: 25645874
26.
Kauffman SA. Metabolic stability and epigenesis in randomly constructed genetic nets. J Theor Biol.
1969; 22: 437–467. PMID: 5803332
27.
Fox RF. Review of Stuart Kauffman, The Origins of Order: Self-Organization and Selection in Evolution.
Biophys J. 1993; 65: 2698–2699.
28.
Irurzun-Arana I, Pastor JM, Troco´niz IF, Go´mez-Mantilla JD. Advanced Boolean modeling of biological
networks applied to systems pharmacology. Bioinformatics. 2017; https://doi.org/10.1093/
bioinformatics/btw747 PMID: 28073755
29.
Naldi A, Monteiro PT, Mu¨ssel C, Consortium for Logical Models and Tools, Kestler HA, Thieffry D, et al.
Cooperative development of logical modelling standards and tools with CoLoMoTo. Bioinformatics.
2015; 31: 1154–1159. https://doi.org/10.1093/bioinformatics/btv013 PMID: 25619997
30.
Abou-Jaoude´ W, Traynard P, Monteiro PT, Saez-Rodriguez J, Helikar T, Thieffry D, et al. Logical
Modeling and Dynamical Analysis of Cellular Networks. Front Genet. 2016; 7: 94. https://doi.org/10.
3389/fgene.2016.00094 PMID: 27303434
31.
Mu¨ssel C, Hopfensitz M, Kestler HA. BoolNet—an R package for generation, reconstruction and analy-
sis of Boolean networks. Bioinformatics. Oxford University Press; 2010; 26: 1378–1380. https://doi.org/
10.1093/bioinformatics/btq124 PMID: 20378558
32.
Chaouiya C, Naldi A, Thieffry D. Logical Modelling of Gene Regulatory Networks with GINsim. In: van
Helden J, Toussaint A, Thieffry D, editors. Bacterial Molecular Networks: Methods and Protocols. New
York, NY: Springer New York; 2012. pp. 463–479.
33.
Mei Y, Abedi V, Carbo A, Zhang X, Lu P, Philipson C, et al. Multiscale modeling of mucosal immune
responses. BMC Bioinformatics. 2015; 16 Suppl 12: S2.
34.
Bassaganya-Riera J. Computational Immunology: Models and Tools. Academic Press; 2015.
35.
Chaouiya C, Be´renguier D, Keating SM, Naldi A, van Iersel MP, Rodriguez N, et al. SBML qualitative
models: a model representation format and infrastructure to foster interactions between qualitative
modelling formalisms and tools. BMC Syst Biol. 2013; 7: 135. https://doi.org/10.1186/1752-0509-7-135
PMID: 24321545
36.
Violeta Balbas-Martinez, Leire Ruiz-Cerda´, Itziar Irurzun-Arana, Ignacio Gonza´lez-Garcı´a, Jose´ David
Go´mez-Mantilla and Iñaki F. Troco´niz. Systems Pharmacology model for Inflammatory Bowel Disease
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
16 / 19
(IBD). In: International Conference on Systems Biology 2016, editor. https://doi.org/10.3252/pso.eu.
17ICSB.2016
37.
Smits HH, van Beelen AJ, Hessle C, Westland R, de Jong E, Soeteman E, et al. Commensal Gram-
negative bacteria prime human dendritic cells for enhanced IL-23 and IL-27 expression and enhanced
Th1 development. Eur J Immunol. 2004; 34: 1371–1380. https://doi.org/10.1002/eji.200324815 PMID:
15114670
38.
Inohara N, Ogura Y, Fontalba A, Gutierrez O, Pons F, Crespo J, et al. Host recognition of bacterial mur-
amyl dipeptide mediated through NOD2. Implications for Crohn’s disease. J Biol Chem. 2003; 278:
5509–5512. https://doi.org/10.1074/jbc.C200673200 PMID: 12514169
39.
Wehkamp J, Frick J-S. Microbiome and chronic inflammatory bowel diseases. J Mol Med. 2016; https://
doi.org/10.1007/s00109-016-1495-z PMID: 27988792
40.
Butto´ LF, Haller D. Dysbiosis in intestinal inflammation: Cause or consequence. Int J Med Microbiol.
2016; https://doi.org/10.1016/j.ijmm.2016.02.010 PMID: 27012594
41.
Glasser A-L, Darfeuille-Michaud A. Abnormalities in the handling of intracellular bacteria in Crohn’s dis-
ease: a link between infectious etiology and host genetic susceptibility. Arch Immunol Ther Exp. 2008;
56: 237–244.
42.
Biancheri P, Di Sabatino A, Corazza GR, MacDonald TT. Proteases and the gut barrier. Cell Tissue
Res. 2013; 351: 269–280. https://doi.org/10.1007/s00441-012-1390-z PMID: 22427120
43.
Pender SLF, MacDonald TT. Matrix metalloproteinases and the gut—new roles for old enzymes. Curr
Opin Pharmacol. 2004; 4: 546–550. https://doi.org/10.1016/j.coph.2004.06.005 PMID: 15525541
44.
Bamba S, Andoh A, Yasui H, Araki Y, Bamba T, Fujiyama Y. Matrix metalloproteinase-3 secretion from
human colonic subepithelial myofibroblasts: role of interleukin-17. J Gastroenterol. 2003; 38: 548–554.
https://doi.org/10.1007/s00535-002-1101-8 PMID: 12825130
45.
Lawrance IC, Rogler G, Bamias G, Breynaert C, Florholmen J, Pellino G, et al. Cellular and Molecular
Mediators of Intestinal Fibrosis. J Crohns Colitis. 2015; https://doi.org/10.1016/j.crohns.2014.09.008
PMID: 25306501
46.
Geremia A, Biancheri P, Allan P, Corazza GR, Di Sabatino A. Innate and adaptive immunity in inflam-
matory bowel disease. Autoimmun Rev. 2014; 13: 3–10. https://doi.org/10.1016/j.autrev.2013.06.004
PMID: 23774107
47.
O’Sullivan S, Gilmer JF, Medina C. Matrix metalloproteinases in inflammatory bowel disease: an
update. Mediators Inflamm. 2015; 2015: 964131. https://doi.org/10.1155/2015/964131 PMID:
25948887
48.
Monteleone G, Caruso R, Fina D, Peluso I, Gioia V, Stolfi C, et al. Control of matrix metalloproteinase
production in human intestinal fibroblasts by interleukin 21. Gut. 2006; 55: 1774–1780. https://doi.org/
10.1136/gut.2006.093187 PMID: 16682426
49.
Helikar T, Kowal B, Rogers JA. A cell simulator platform: the cell collective. Clin Pharmacol Ther. 2013;
93: 393–395. https://doi.org/10.1038/clpt.2013.41 PMID: 23549147
50.
Helikar T, Kowal B, McClenathan S, Bruckner M, Rowley T, Madrahimov A, et al. The Cell Collective:
toward an open and collaborative approach to systems biology. BMC Syst Biol. 2012; 6: 96. https://doi.
org/10.1186/1752-0509-6-96 PMID: 22871178
51.
Boldhaus G, Bertschinger N, Rauh J, Olbrich E, Klemm K. Robustness of Boolean dynamics under
knockouts. Phys Rev E Stat Nonlin Soft Matter Phys. 2010; 82: 021916. https://doi.org/10.1103/
PhysRevE.82.021916 PMID: 20866846
52.
Taylor AL, Watson CJE, Bradley JA. Immunosuppressive agents in solid organ transplantation: Mecha-
nisms of action and therapeutic efficacy. Crit Rev Oncol Hematol. 2005; 56: 23–46. https://doi.org/10.
1016/j.critrevonc.2005.03.012 PMID: 16039869
53.
McManus R. Mechanisms of steroid action and resistance in inflammation and disease. J Endocrinol.
2003; 178: 1–4. PMID: 12844329
54.
Coutinho AE, Chapman KE. The anti-inflammatory and immunosuppressive effects of glucocorticoids,
recent developments and mechanistic insights. Mol Cell Endocrinol. 2011; 335: 2–13. https://doi.org/10.
1016/j.mce.2010.04.005 PMID: 20398732
55.
Peters LA, Perrigoue J, Mortha A, Iuga A, Song W-M, Neiman EM, et al. A functional genomics predic-
tive network model identifies regulators of inflammatory bowel disease. Nat Genet. 2017; 49: 1437–
1449. https://doi.org/10.1038/ng.3947 PMID: 28892060
56.
Laffin M, Madsen KL. Fecal Microbial Transplantation in Inflammatory Bowel Disease: A Movement
Too Big to Be Ignored. Clin Pharmacol Ther. 2017; 102: 588–590. https://doi.org/10.1002/cpt.747
PMID: 28695658
57.
Fischer S, Neurath MF. Precision Medicine in Inflammatory Bowel Diseases. Clin Pharmacol Ther.
2017; 102: 623–632. https://doi.org/10.1002/cpt.793 PMID: 28699158
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
17 / 19
58.
Dipasquale V, Romano C. Vaccination strategies in pediatric inflammatory bowel disease. Vaccine.
2017; https://doi.org/10.1016/j.vaccine.2017.09.031 PMID: 28967524
59.
Danese S, Furfaro F, Vetrano S. Targeting S1P in Inflammatory bowel disease: new avenues for modu-
lating intestinal leukocyte migration. J Crohns Colitis. 2017; https://doi.org/10.1093/ecco-jcc/jjx107
PMID: 28961752
60.
Fengming Y, Jianbing W. Biomarkers of inflammatory bowel disease. Dis Markers. 2014; 2014:
710915. https://doi.org/10.1155/2014/710915 PMID: 24963213
61.
Helikar T, Konvalina J, Heidel J, Rogers JA. Emergent decision-making in biological signal transduction
networks. Proc Natl Acad Sci U S A. 2008; 105: 1913–1918. https://doi.org/10.1073/pnas.0705088105
PMID: 18250321
62.
Ben-Horin S, Vande Casteele N, Schreiber S, Lakatos PL. Biosimilars in Inflammatory Bowel Disease:
Facts and Fears of Extrapolation. Clin Gastroenterol Hepatol. 2016; https://doi.org/10.1016/j.cgh.2016.
05.023 PMID: 27215364
63.
van Dullemen HM, van Deventer SJ, Hommes DW, Bijl HA, Jansen J, Tytgat GN, et al. Treatment of
Crohn’s disease with anti-tumor necrosis factor chimeric monoclonal antibody (cA2). Gastroenterology.
1995; 109: 129–135. PMID: 7797011
64.
Present DH, Rutgeerts P, Targan S, Hanauer SB, Mayer L, van Hogezand RA, et al. Infliximab for the
treatment of fistulas in patients with Crohn’s disease. N Engl J Med. 1999; 340: 1398–1405. https://doi.
org/10.1056/NEJM199905063401804 PMID: 10228190
65.
Colombel J-F, Sandborn WJ, Rutgeerts P, Enns R, Hanauer SB, Panaccione R, et al. Adalimumab for
maintenance of clinical response and remission in patients with Crohn’s disease: the CHARM trial.
Gastroenterology. 2007; 132: 52–65. https://doi.org/10.1053/j.gastro.2006.11.041 PMID: 17241859
66.
Yoshimura N, Yokoyama Y, Matsuoka K, Takahashi H, Iwakiri R, Yamamoto T, et al. An open-label pro-
spective randomized multicenter study of intensive versus weekly granulocyte and monocyte apheresis
in active crohn’s disease. BMC Gastroenterol. 2015; 15: 163. https://doi.org/10.1186/s12876-015-
0390-3 PMID: 26585569
67.
Liu Z, Jiang X, Sun C. The efficacy and safety of selective granulocyte and monocyte apheresis for
inflammatory bowel disease: A meta-analysis. Eur J Intern Med. 2016; 36: e26–e27. https://doi.org/10.
1016/j.ejim.2016.08.028 PMID: 27614377
68.
Di Girolamo M, Sartini A, Critelli R, Bertani A, Merighi A, Villa E. Efficacy of a Novel Granulocyte Mono-
cyte Apheresis Adsorber Device in the Treatment of Inflammatory Bowel Diseases: A Pilot Study. Ther
Apher Dial. 2016; 20: 668–676. https://doi.org/10.1111/1744-9987.12453 PMID: 27921374
69.
Hommes DW, Mikhajlova TL, Stoinov S, Stimac D, Vucelic B, Lonovics J, et al. Fontolizumab, a huma-
nised anti-interferon gamma antibody, demonstrates safety and clinical activity in patients with moder-
ate to severe Crohn’s disease. Gut. 2006; 55: 1131–1137. https://doi.org/10.1136/gut.2005.079392
PMID: 16507585
70.
Reinisch W, Hommes DW, Van Assche G, Colombel J-F, Gendre J-P, Oldenburg B, et al. A dose esca-
lating, placebo controlled, double blind, single dose and multidose, safety and tolerability study of fonto-
lizumab, a humanised anti-interferon gamma antibody, in patients with moderate to severe Crohn’s
disease. Gut. 2006; 55: 1138–1144. https://doi.org/10.1136/gut.2005.079434 PMID: 16492717
71.
Schreiber S, Fedorak RN, Nielsen OH, Wild G, Williams CN, Nikolaus S, et al. Safety and efficacy of
recombinant human interleukin 10 in chronic active Crohn’s disease. Crohn’s Disease IL-10 Coopera-
tive Study Group. Gastroenterology. 2000; 119: 1461–1472. PMID: 11113067
72.
Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, Reinisch W, Higgins PDR, et al. Secukinu-
mab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected
results of a randomised, double-blind placebo-controlled trial. Gut. 2012; 61: 1693–1700. https://doi.
org/10.1136/gutjnl-2011-301668 PMID: 22595313
73.
Sands BE, Sandborn WJ, Creed TJ, Dayan CM, Dhanda AD, Van Assche GA, et al. Basiliximab does
not increase efficacy of corticosteroids in patients with steroid-refractory ulcerative colitis. Gastroenter-
ology. 2012; 143: 356–64.e1. https://doi.org/10.1053/j.gastro.2012.04.043 PMID: 22549092
74.
Van Assche G, Sandborn WJ, Feagan BG, Salzberg BA, Silvers D, Monroe PS, et al. Daclizumab, a
humanised monoclonal antibody to the interleukin 2 receptor (CD25), for the treatment of moderately to
severely active ulcerative colitis: a randomised, double blind, placebo controlled, dose ranging trial. Gut.
BMJ Publishing Group Ltd and British Society of Gastroenterology; 2006; 55: 1568–1574. https://doi.
org/10.1136/gut.2005.089854 PMID: 16603634
75.
Paliogianni F, Ahuja SS, Balow JP, Balow JE, Boumpas DT. Novel mechanism for inhibition of human T
cells by glucocorticoids. Glucocorticoids inhibit signal transduction through IL-2 receptor. J Immunol.
1993; 151: 4081–4089. PMID: 8409387
76.
Horst HJ, Flad HD. Corticosteroid-interleukin 2 interactions: inhibition of binding of interleukin 2 to inter-
leukin 2 receptors. Clin Exp Immunol. 1987; 68: 156–161. PMID: 3115640
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
18 / 19
77.
El Menyawi M, Fawzy M, Al-Nahas Z, Edris A, Hussein H, Shaker O, et al. Serum tumor necrosis factor
alpha (TNF-α) level in patients with Behc¸et’s disease: Relation to clinical manifestations and disease
activity. The Egyptian Rheumatologist. 2014; 36: 139–143.
78.
Shi M, Wei J, Dong J, Meng W, Ma J, Wang T, et al. Function of interleukin-17 and -35 in the blood of
patients with hepatitis B-related liver cirrhosis. Mol Med Rep. 2015; 11: 121–126. https://doi.org/10.
3892/mmr.2014.2681 PMID: 25323532
79.
Thomas DW, Burns J, Audette J, Carrol A, Dow-Hygelund C, Hay M. Clinical development success
rates 2006–2015. San Diego: Biomedtracker/Washington, DC: BIO/Bend: Amplion. 2016;
80.
Furfaro F, Gilardi D, Allocca M, Cicerone C, Correale C, Fiorino G, et al. IL-23 Blockade for Crohn s dis-
ease: next generation of anti-cytokine therapy. Expert Rev Clin Immunol. 2017; 1–11.
81.
Palsson S, Hickling TP, Bradshaw-Pierce EL, Zager M, Jooss K, O’Brien PJ, et al. The development of
a fully-integrated immune response model (FIRM) simulator of the immune response through integra-
tion of multiple subset models. BMC Syst Biol. 2013; 7: 95. https://doi.org/10.1186/1752-0509-7-95
PMID: 24074340
82.
Carbo A, Hontecillas R, Kronsteiner B, Viladomiu M, Pedragosa M, Lu P, et al. Systems modeling of
molecular mechanisms controlling cytokine-driven CD4+ T cell differentiation and phenotype plasticity.
PLoS Comput Biol. 2013; 9: e1003027. https://doi.org/10.1371/journal.pcbi.1003027 PMID: 23592971
83.
Laurence A O’Shea JJ. TH-17 differentiation: of mice and men. Nat Immunol. Nature Publishing Group;
2007; 8: 903–905. https://doi.org/10.1038/ni0907-903 PMID: 17712339
84.
Reynolds G, Haniffa M. Human and Mouse Mononuclear Phagocyte Networks: A Tale of Two Species?
Front Immunol. 2015; 6: 330. https://doi.org/10.3389/fimmu.2015.00330 PMID: 26124761
85.
Mestas J, Hughes CCW. Of mice and not men: differences between mouse and human immunology. J
Immunol. 2004; 172: 2731–2738. PMID: 14978070
86.
Wasilewska A, Winiarska M, Olszewska M, Rudnicka L. Interleukin-17 inhibitors. A new era in treatment
of psoriasis and other skin diseases. Postepy Dermatol Alergol. 2016; 33: 247–252. https://doi.org/10.
5114/ada.2016.61599 PMID: 27605893
87.
Tan Y, Qi Q, Lu C, Niu X, Bai Y, Jiang C, et al. Cytokine Imbalance as a Common Mechanism in Both
Psoriasis and Rheumatoid Arthritis. Mediators Inflamm. 2017; 2017: 2405291. https://doi.org/10.1155/
2017/2405291 PMID: 28239238
88.
Ryll A, Samaga R, Schaper F, Alexopoulos LG, Klamt S. Large-scale network models of IL-1 and IL-6
signalling and their hepatocellular specification. Mol Biosyst. 2011; 7: 3253–3270. https://doi.org/10.
1039/c1mb05261f PMID: 21968890
89.
Kauffman SA. Metabolic stability and epigenesis in randomly constructed genetic nets. J Theor Biol.
1969; 22: 437–467. PMID: 5803332
90.
Kauffman SA. The origins of order: Self organization and selection in evolution. Oxford University
Press, USA; 1993.
91.
Hopfensitz M, Mu¨ssel C, Maucher M, Kestler HA. Attractors in Boolean networks: a tutorial. Comput
Stat. Springer-Verlag; 2012; 28: 19–36.
92.
Saadatpour A, Albert I, Albert R. Attractor analysis of asynchronous Boolean models of signal transduc-
tion networks. J Theor Biol. 2010; 266: 641–656. https://doi.org/10.1016/j.jtbi.2010.07.022 PMID:
20659480
93.
Wynn ML, Consul N, Merajver SD, Schnell S. Logic-based models in systems biology: a predictive and
parameter-free network analysis method. Integr Biol. 2012; 4: 1323–1337.
94.
Saadatpour A, Albert I, Albert R. Attractor analysis of asynchronous Boolean models of signal transduc-
tion networks. J Theor Biol. Elsevier; 2010; 266: 641–656. https://doi.org/10.1016/j.jtbi.2010.07.022
PMID: 20659480
95.
Thakar J, Pilione M, Kirimanjeswara G, Harvill ET, Albert R. Modeling systems-level regulation of host
immune responses. PLoS Comput Biol. 2007; 3: e109. https://doi.org/10.1371/journal.pcbi.0030109
PMID: 17559300
96.
Harvey I, Bossomaier T. Time out of joint: Attractors in asynchronous random boolean networks. Pro-
ceedings of the Fourth European Conference on Artificial Life. MIT Press, Cambridge; 1997. pp. 67–75.
97.
Willadsen K, Triesch J, Wiles J. Understanding robustness in Random Boolean Networks. ALIFE.
2008. pp. 694–701.
98.
Rokach L, Maimon O. Clustering Methods. In: Maimon O, Rokach L, editors. Data Mining and Knowl-
edge Discovery Handbook. Springer US; 2005. pp. 321–352.
A systems pharmacology model for IBD
PLOS ONE | https://doi.org/10.1371/journal.pone.0192949
March 7, 2018
19 / 19
|
29513758
|
PERFOR = ( NK_NKG2D ) OR ( NK )
Th2upregulation = ( Th2 AND ( ( ( IL4 ) ) ) )
IL15 = ( MACR AND ( ( ( LPS OR IFNg ) ) ) ) OR ( FIBROBLAST AND ( ( ( PGN OR LPS OR MDP ) ) ) )
IL23 = ( MACR AND ( ( ( IL1b ) ) ) ) OR ( DC )
CD8_NKG2D = ( ( MDP ) AND NOT ( CD8_NKG2D AND ( ( ( IL2 AND IL21 ) ) OR ( ( IEC_ULPB1_6 ) ) OR ( ( IEC_MICA_B ) ) ) ) ) OR ( ( LPS ) AND NOT ( CD8_NKG2D AND ( ( ( IL2 AND IL21 ) ) OR ( ( IEC_ULPB1_6 ) ) OR ( ( IEC_MICA_B ) ) ) ) ) OR ( ( PGN ) AND NOT ( CD8_NKG2D AND ( ( ( IL2 AND IL21 ) ) OR ( ( IEC_ULPB1_6 ) ) OR ( ( IEC_MICA_B ) ) ) ) )
Th2 = ( ( Th0 AND ( ( ( IL4 AND IL18 ) AND ( ( ( NOT IL12 ) ) ) ) OR ( ( IL10 ) ) OR ( ( Th2 AND IL4 ) AND ( ( ( NOT Th2upregulation ) ) ) ) ) ) AND NOT ( Th2 AND ( ( ( Treg OR TGFb OR IFNg ) ) ) ) )
Treg = ( ( Th0 AND ( ( ( TLR2 OR TGFb ) ) ) ) AND NOT ( Treg AND ( ( ( IL23 OR IL6 OR IL21 OR IL22 OR Th17 OR TNFa ) ) ) ) )
IL1b = ( ( MACR AND ( ( ( NFkB AND LPS ) ) ) ) AND NOT ( IL10 AND ( ( ( IL1b ) ) ) ) ) OR ( ( DC AND ( ( ( NFkB AND LPS ) ) ) ) AND NOT ( IL10 AND ( ( ( IL1b ) ) ) ) )
TLR2 = ( PGN )
Th17 = ( ( Th0 AND ( ( ( IL23 OR IL6 OR IL1b ) ) ) ) AND NOT ( Th17 AND ( ( ( IL4 OR Treg OR IL12 OR TGFb OR IFNg ) ) ) ) )
IL6 = ( MACR AND ( ( ( PGN ) ) ) ) OR ( Th17 AND ( ( ( IL23 ) ) ) ) OR ( NFkB AND ( ( ( NOT IL10 OR NOT IL4 ) ) ) ) OR ( DC AND ( ( ( PGN OR LPS ) ) ) )
IEC_ULPB1_6 = ( CD8_NKG2D AND ( ( ( PGN OR LPS OR MDP ) ) ) )
FIBROBLAST = ( ( IL2 ) AND NOT ( FIBROBLAST AND ( ( ( IL12 OR IFNg ) ) ) ) ) OR ( ( MACR AND ( ( ) OR ( ( IL4 OR IL13 OR TGFb ) ) ) ) AND NOT ( FIBROBLAST AND ( ( ( IL12 OR IFNg ) ) ) ) )
NFkB = ( TLR4 ) OR ( NOD2 ) OR ( TLR2 )
NK = ( ( IL18 AND ( ( ( IL10 ) ) ) ) AND NOT ( NK AND ( ( ( Treg ) ) ) ) ) OR ( ( IL23 ) AND NOT ( NK AND ( ( ( Treg ) ) ) ) ) OR ( ( DC AND ( ( ( IL15 ) ) ) ) AND NOT ( NK AND ( ( ( Treg ) ) ) ) )
Th0_M = ( Th0 AND ( ( ( IL23 OR IL12 ) ) ) ) OR ( Th0_M )
LPS = NOT ( ( DEF ) OR ( GRANZB ) OR ( PERFOR ) )
NK_NKG2D = ( ( MDP ) AND NOT ( NK_NKG2D AND ( ( ( IL21 ) AND ( ( ( IL12 ) ) ) ) AND ( ( IEC_ULPB1_6 ) ) AND ( ( IEC_MICA_B ) ) AND ( ( TGFb ) ) ) ) ) OR ( ( PGN ) AND NOT ( NK_NKG2D AND ( ( ( IL21 ) AND ( ( ( IL12 ) ) ) ) AND ( ( IEC_ULPB1_6 ) ) AND ( ( IEC_MICA_B ) ) AND ( ( TGFb ) ) ) ) ) OR ( ( LPS ) AND NOT ( NK_NKG2D AND ( ( ( IL21 ) AND ( ( ( IL12 ) ) ) ) AND ( ( IEC_ULPB1_6 ) ) AND ( ( IEC_MICA_B ) ) AND ( ( TGFb ) ) ) ) )
CD4_NKG2Dupregulation = ( CD4_NKG2D AND ( ( ( IL15 OR TNFa ) ) ) )
MMPs = ( MACR AND ( ( ( TNFa ) ) ) ) OR ( FIBROBLAST AND ( ( ( IL21 OR IL17 OR TNFa OR IL1b ) ) ) )
IEC_MICA_B = ( ( MDP ) AND NOT ( TGFb ) ) OR ( ( LPS ) AND NOT ( TGFb ) ) OR ( ( IEC_MICA_B AND ( ( ( TNFa ) AND ( ( ( NOT IEC_MICA_Bupregulation ) ) ) ) ) ) AND NOT ( TGFb ) ) OR ( ( PGN ) AND NOT ( TGFb ) )
DEF = ( IL22 ) OR ( NOD2 ) OR ( IL17 )
CD4_NKG2D = ( ( MDP ) AND NOT ( CD4_NKG2D AND ( ( ( IL10 ) ) OR ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) ) OR ( ( LPS ) AND NOT ( CD4_NKG2D AND ( ( ( IL10 ) ) OR ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) ) OR ( ( PGN ) AND NOT ( CD4_NKG2D AND ( ( ( IL10 ) ) OR ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) ) OR ( ( CD4_NKG2D AND ( ( ( IL15 OR TNFa ) AND ( ( ( NOT CD4_NKG2Dupregulation ) ) ) ) ) ) AND NOT ( CD4_NKG2D AND ( ( ( IL10 ) ) OR ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) )
IL12 = ( LPS AND ( ( ( IFNg ) AND ( ( ( DC ) ) OR ( ( PGN AND MACR ) ) ) ) ) ) OR ( TLR2 AND ( ( ( NFkB ) AND ( ( ( MACR OR DC ) ) ) ) ) )
IL21 = ( ( ( ( Th0 AND ( ( ( IL6 ) ) ) ) AND NOT ( IFNg ) ) AND NOT ( TGFb ) ) AND NOT ( IL4 ) ) OR ( Th17 )
IEC_MICA_Bupregulation = ( IEC_MICA_B AND ( ( ( TNFa ) ) ) )
DC = ( ( TLR4 ) AND NOT ( DC AND ( ( ( IL10 ) ) ) ) ) OR ( ( TLR2 ) AND NOT ( DC AND ( ( ( IL10 ) ) ) ) ) OR ( ( NOD2 ) AND NOT ( DC AND ( ( ( IL10 ) ) ) ) )
IFNg = ( ( ( IL18 AND ( ( ( IL12 ) AND ( ( ( Th0 OR MACR ) ) ) ) ) ) AND NOT ( IFNg AND ( ( ( IL10 OR TGFb ) ) ) ) ) AND NOT ( Th2 ) ) OR ( ( ( NK_NKG2D AND ( ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) AND NOT ( IFNg AND ( ( ( IL10 OR TGFb ) ) ) ) ) AND NOT ( Th2 ) ) OR ( ( ( CD8_NKG2D AND ( ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) AND NOT ( IFNg AND ( ( ( IL10 OR TGFb ) ) ) ) ) AND NOT ( Th2 ) ) OR ( ( ( Th1 ) AND NOT ( IFNg AND ( ( ( IL10 OR TGFb ) ) ) ) ) AND NOT ( Th2 ) ) OR ( ( ( Th17 AND ( ( ( PGN OR LPS OR MDP ) ) ) ) AND NOT ( IFNg AND ( ( ( IL10 OR TGFb ) ) ) ) ) AND NOT ( Th2 ) ) OR ( ( ( IL23 AND ( ( ( NK ) ) AND ( ( PGN OR LPS OR MDP ) ) ) ) AND NOT ( IFNg AND ( ( ( IL10 OR TGFb ) ) ) ) ) AND NOT ( Th2 ) )
TNFa = ( ( MACR AND ( ( ( LPS AND IFNg ) ) OR ( ( PGN ) ) OR ( ( IL2 ) ) ) ) AND NOT ( IL10 AND ( ( ( TNFa ) AND ( ( ( TLR4 OR TLR2 ) ) ) ) ) ) ) OR ( ( NFkB AND ( ( ( LPS ) ) ) ) AND NOT ( IL10 AND ( ( ( TNFa ) AND ( ( ( TLR4 OR TLR2 ) ) ) ) ) ) ) OR ( ( NK_NKG2D AND ( ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) AND NOT ( IL10 AND ( ( ( TNFa ) AND ( ( ( TLR4 OR TLR2 ) ) ) ) ) ) ) OR ( ( CD8_NKG2D AND ( ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) AND NOT ( IL10 AND ( ( ( TNFa ) AND ( ( ( TLR4 OR TLR2 ) ) ) ) ) ) ) OR ( ( NK AND ( ( ( PGN OR LPS OR MDP ) AND ( ( ( IL23 OR IL2 OR IL15 ) ) ) ) ) ) AND NOT ( IL10 AND ( ( ( TNFa ) AND ( ( ( TLR4 OR TLR2 ) ) ) ) ) ) ) OR ( ( FIBROBLAST AND ( ( ( IFNg ) ) ) ) AND NOT ( IL10 AND ( ( ( TNFa ) AND ( ( ( TLR4 OR TLR2 ) ) ) ) ) ) ) OR ( ( CD4_NKG2D AND ( ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) AND NOT ( IL10 AND ( ( ( TNFa ) AND ( ( ( TLR4 OR TLR2 ) ) ) ) ) ) )
IL13 = ( Th2 )
IL18 = ( LPS AND ( ( ( MACR OR DC ) ) AND ( ( NFkB ) ) ) )
TLR4 = ( LPS )
NOD2 = ( MDP )
Th1 = ( ( Th0 AND ( ( ( IL18 OR IL12 OR IFNg ) ) ) ) AND NOT ( Th1 AND ( ( ( IL10 ) ) OR ( ( Treg ) ) OR ( ( IL12 ) AND ( ( ( IL23 OR IL17 ) ) ) ) OR ( ( TGFb ) ) OR ( ( Th2 ) ) OR ( ( IL4 ) ) ) ) )
IL22upregulation = ( Th0 AND ( ( ( IL21 ) ) AND ( ( IL22 ) ) ) )
IL10 = ( MACR AND ( ( ( LPS ) AND ( ( ( NOT IL4 ) ) ) ) ) ) OR ( DC AND ( ( ( LPS ) ) ) ) OR ( TLR2 AND ( ( ( NFkB ) AND ( ( ( NOT MACR AND NOT IFNg ) ) ) ) ) ) OR ( Th2 AND ( ( ( NOT IL23 ) ) ) ) OR ( Treg )
PGN = NOT ( ( DEF ) OR ( PERFOR ) OR ( GRANZB ) )
MACR = ( ( NOD2 ) AND NOT ( MACR AND ( ( ( IL10 ) ) ) ) ) OR ( ( IFNg ) AND NOT ( MACR AND ( ( ( IL10 ) ) ) ) ) OR ( ( IL15 ) AND NOT ( MACR AND ( ( ( IL10 ) ) ) ) ) OR ( ( TLR4 ) AND NOT ( MACR AND ( ( ( IL10 ) ) ) ) ) OR ( ( TLR2 ) AND NOT ( MACR AND ( ( ( IL10 ) ) ) ) )
TGFb = ( MACR ) OR ( Treg )
GRANZB = ( CD8_NKG2D ) OR ( NK ) OR ( DC AND ( ( ( NOT PGN OR NOT LPS ) ) ) ) OR ( NK_NKG2D )
MDP = NOT ( ( DEF ) OR ( GRANZB ) OR ( PERFOR ) )
IL22 = ( NK AND ( ( ( IL18 AND IL12 ) ) ) ) OR ( Th17 ) OR ( ( Th0 AND ( ( ( IL21 ) ) AND ( ( NOT IL22upregulation ) ) AND ( ( IL22 ) ) ) ) AND NOT ( TGFb ) ) OR ( CD4_NKG2D )
Th17_M = ( Th17_M ) OR ( Th0_M AND ( ( ( PGN OR LPS OR MDP ) AND ( ( ( IL2 ) ) OR ( ( IL6 AND IL1b ) ) OR ( ( IL23 ) ) ) ) ) )
IL4 = ( Th2 )
IL17 = ( ( Th17 ) AND NOT ( IL17 AND ( ( ( IL13 OR TGFb ) ) ) ) ) OR ( ( CD4_NKG2D AND ( ( ( IEC_ULPB1_6 OR IEC_MICA_B ) ) ) ) AND NOT ( IL17 AND ( ( ( IL13 OR TGFb ) ) ) ) ) OR ( ( Th17_M AND ( ( ( PGN OR LPS OR MDP ) ) ) ) AND NOT ( IL17 AND ( ( ( IL13 OR TGFb ) ) ) ) )
IL2 = ( Th0_M AND ( ( ( PGN OR LPS OR MDP ) ) ) ) OR ( Th0 ) OR ( DC )
Th0 = ( MDP ) OR ( PGN ) OR ( LPS )
|
Subsets and Splits
No community queries yet
The top public SQL queries from the community will appear here once available.