Abstract
Cellular identity as defined through morphology and function emerges from intracellular signaling networks that communicate between cells. Based on recursive interactions within and among these intracellular networks, dynamical solutions in terms of biochemical behavior are generated that can differ from those in isolated cells. In this way, cellular heterogeneity in tissues can be established, implying that cell identity is not intrinsically predetermined by the genetic code but is rather dynamically maintained in a cognitive manner. We address how to experimentally measure the flow of information in intracellular biochemical networks and demonstrate that even simple causality motifs can give rise to rich, context‐dependent dynamic behavior. The concept how intercellular communication can result in novel dynamical solutions is applied to provide a contextual perspective on cell differentiation and tumorigenesis.
Keywords: cellular fate, intercellular communication, network reconstruction, network topology, protein network dynamics
Subject Categories: Signal Transduction, Systems & Computational Biology
Introduction
Cells require encapsulation to spatially organize their chemical constituents such that the time and place of molecular interactions are a necessary determinant of their effect (Farnsworth et al, 2013). Under such confinement, they can control the flow of matter and energy, thereby maintaining themselves in entropy‐dissipating non‐equilibrium conditions (Schrödinger, 1944; Prigogine & Nicolis, 1967). This enables information to be concentrated in cells in a form of self‐organizing chemical activity patterns (Turing, 1952; Grecco et al, 2011). The cells continuously adjust these internal states to allow for robustness in identity under changing external conditions, especially to unfamiliar changes for which no regulatory program has been fixed in advance (Ibarra et al, 2002; Fong & Palsson, 2004; Kashiwagi et al, 2006; Stolovicki et al, 2006). Such flexible dynamics therefore implies that genes serve as repositories of dynamic control information regarding the combination of interacting proteins that define a stable phenotypic state. Distinct biochemical manifestations of this information pool reflect proteome plasticity: In multicellular organisms, all cells contain nearly identical copies of the genome but exhibit drastically different phenotypes. This principle also accounts for efficient adaptation on evolutionary as well as short timescales, and implicitly incorporates the “tinkering” aspect of biological systems (Jacob, 1977). Cells thereby define a wider dynamical domain in which they can interact with their environment to generate and maintain their identity, a process reminiscent to cognition (Box 1; Maturana & Varela, 1980). In a very general formulation, cognition can be defined as a set of processes that are in a structural coupling with the environment such that the system adapts to its environment or transforms the environment to adapt it to the “needs” of the system (Bourgine & Stewart, 2004). In terms of living systems, cognition is realized via recursive chemical communication of intracellular signaling networks between cells. Thus, processing external chemical cues on the level of intracellular networks, and at the same time emitting the information about their state to the neighboring cells, allows cellular entities in tissues to establish a collective behavior to maintain themselves. Depending on the conditions, the behavior of this network of signaling networks converges to one of multiple stable dynamical solutions that can differ from those in isolated cells.
Box 1: Cellular cognition.
In the 1980 book “Autopoiesis and Cognition—the realization of the living”, H. Maturana and F. Varela defined living systems as “… units of interactions, they exist in ambience. From a purely biological point of view, they can not be understood independently of the part of the ambience with which they interact: the niche; nor can the niche be defined independently of the living system that specifies it” (Maturana & Varela, 1980). On the basis of this concept, Maturana then defined cognition as a process driven by structural, bidirectional causal organism–environment relations that are necessary to understand coding and information transmission in living systems. In 1993, J. Stewart related the concept of cellular cognition to the characteristics of the immune system within a living organism in order to encompass its properties of recognition, learning, memory, and self/non‐self discrimination into one theoretical framework (Stewart, 1993). He discussed these properties in relation to neuronal systems and higher‐order cognition. Later on, Bourgine and Stewart proposed that living systems must be both autopoietic and cognitive, providing more refined definitions for these concepts. In their view, an autopoietic system is “a network of processes that produces the components that reproduce the network, and also regulates the boundary conditions necessary for its ongoing existence as a network”. Moreover, “A system is cognitive if and only if sensory inputs serve to trigger actions in a specific way, so as to satisfy a viability constraint” (Bourgine & Stewart, 2004).
Cells therefore necessitate both autopoiesis and cognition (Maturana & Varela, 1980) to generate and dynamically maintain heterogeneous cellular identities in tissues. Autopoiesis is a necessary characteristic of living systems to replicate the components and reproduce the architectures of biochemical networks, thereby maintaining themselves and the boundary conditions necessary for their own existence (Bourgine & Stewart, 2004). Cognition on the other hand is required to differentiate the replicated entities through the dynamics of the network of networks that is established through bidirectional intercellular communication (Fig 1). In this way, cognition is a property that emerges from recursive interactions between the signaling networks of cells.
Based on this framework, we discuss what determines the dynamics of signaling networks and how this information can be extracted experimentally. We start by considering undirected protein interaction networks derived from proteomic approaches. We argue that temporal behavior of the protein reactants is necessary to deduce the causality of intracellular networks and thereby their dynamical potential. From there, we describe how intercellular communication endows the system with cognitive properties, generating new dynamical behavior different than the one in the isolated cells. As an example, we elaborate on a Turing‐like principle that accounts for the emergence of diverse identities in a clonal cell population. In this context, we also discuss how the collective behavior in a normal tissue can be affected by changes in the cognitive abilities of cells induced upon oncogenic mutations. Thus, by considering cellular identity to be dynamically maintained by recursive interactions, we explore whether cells can learn to perceive their environment and thereby change their identity.
What protein interactions tell us about cellular states
Current proteomic approaches allow quantitative detection of protein abundances and protein reactions in terms of protein complexes and post‐translational modifications (PTMs) in ensemble of cells (Cox & Mann, 2011; Larance & Lamond, 2015) (Box 2). The protein abundances provide the composition of the proteome reflecting the gene expression in a particular cell population that is studied in a distinct experimental context. The detection of protein complexes and/or PTMs on the other hand gives access to the basic reactions—the currency of signal transduction—through which the cells process extracellular information. A major advantage of all proteomic approaches is that hundreds to thousands of protein complexes or PTMs can be simultaneously and rapidly analyzed. In case of protein interaction maps, the nodes of the obtained protein interaction networks represent the proteins under study and the links or the edges represent their physical interactions (Gavin et al, 2002; Völkel et al, 2010). These interactions can be both, direct or indirect, since there is no clear separation criterion in the case when several proteins are part of an isolated complex. However, even if all possible PTMs/interactions could be obtained experimentally, this single (static) proteomic snapshot does not provide sufficient information to identify how processing of distinct signals determines the cellular phenotype (Fig 2A). The temporal component is therefore a necessary experimental prerequisite to identify which proteins alter their interactions in response to external stimuli because signals propagate through changes in the reaction states of the network components (Von Kriegsheim et al, 2009; D'Souza et al, 2014; Francavilla et al, 2014; Humphrey et al, 2015).
Box 2: Proteomic approaches to derive protein interactions.
Three main proteomics‐based approaches allow for systematic, large‐scale identification of protein–protein interactions (Larance & Lamond, 2015): (i) methods based on affinity pull‐down where the protein of interest and its interacting partners are specifically isolated. The protein complexes are then eluted for subsequent analysis by digestion and liquid chromatography followed by tandem mass spectrometry (LC‐MS/MS). (ii) Proximity labeling methods in which cell lines are constructed that ectopically express a protein of interest fused to either a promiscuous biotin ligase or a peroxidase enzyme that can further covalently transfer biotin labels to potential interacting proteins in close proximity. LC‐MS/MS methods are further used to identify the corresponding protein interaction map. (iii) Protein interactions can also be identified by protein correlation profiling with chromatography and density gradient centrifugation, for example, to separate endogenous protein complexes according to size, density, charge, or hydrophobicity, assuming that interacting proteins will co‐elute.
The obtained interactions using proteomic approaches can represent both, direct and indirect connections, since it is not always clear whether an identified multiprotein complex is one complex or the summation of different sub‐complexes that contain the same building blocks. By extracting protein complexes from cell lysates, spatial constrains imposed by supramolecular assemblies (like membranes or cytoskeletal structures) on protein complexes are also obliterated. This might result in the loss of complexes that are dependent on the integrity of supramolecular structures as well as generate non‐physiological interactions due to mixing of all components. In addition, information on cell‐to‐cell variability in proteome composition and molecular reactions is lost.
What is relevant to note here is that after killing the cells to extract protein complexes, the derived interaction maps reflect the state of the cell in equilibrium conditions. Even if the fixation of the cells would be so rapid that the proteome cannot relax to equilibrium, the obtained snapshots of the temporal evolution of the reaction states in the proteome represent only one of many possible dynamical solutions of the network. Therefore, no knowledge can be gained how information flows in the network to realize these states. To obtain this level of information, it is necessary to deduce the causal relationships between protein reactions in the proteome.
Causality determines the dynamics of biochemical networks
Causality in biochemical systems is generated when protein reaction states or complexes can affect the interaction or reaction of other proteins. In this manner, directional activatory or inhibitory effects on each other's chemical conversions are established (Fig 2B) (Sachs et al, 2005; Zamir & Bastiaens, 2008). The directionality of the interaction determines how the information flows through the system, processing and translating external cues into specific cellular behavior. This type of representation is referred to as “network topology”, where each node represents a chemical intermediate of a reaction and the directed edges the positive or negative influence of one node on another (Fig 2C).
To illuminate how the topology of molecular networks determines their dynamical behavior (Brandman et al, 2005; Santos et al, 2007; Ryu et al, 2015), we start with the autocatalytic phosphorylation cycle as it typically occurs in receptor tyrosine kinases (Lemmon & Schlessinger, 2010). This reaction cycle has only one stable dynamical solution, which is the phosphorylation steady state of all receptors. This state is dynamically maintained since a constant ATP input is required to sustain the phosphorylation at a certain level, when phosphatase‐mediated hydrolysis removes the phosphates from the tyrosine residues (Fig 3A) (Ostman & Bohmer, 2001). Things become more interesting if the activity of the phosphatase (PTP) is coupled to that of the RTK via Rac‐induced NOX complex activation, which in turn generates reactive oxygen species (ROS), thus inactivating the PTPs (Denu & Tanner, 1998; Finkel, 1998; Meng et al, 2002). This reaction scheme generates a toggle switch—or double‐negative feedback loop—that together with the autocatalytic RTK activity can give rise to bistable behavior (Reynolds et al, 2003) (Fig 3B). In this system, there are three solutions: two dynamically maintained low and high RTK phosphorylation states, both of which are stable and can coexist in a given parameter interval, and an unstable solution that acts as a threshold in the system. In which stable solution the system will end up is directly related to the initial conditions, that is, the initial phosphorylation level of the receptor. In a cellular context, this might be dependent on the history of the cell in terms of prior growth factor‐induced signaling states. When these simple networks interact in a recursive bidirectional way between cells, novel dynamical states can be generated and maintained, different than those of isolated cells. This principle that resembles a cognitive process is further elaborated in the section “Recursive communication between cells leads to cognition”.
On the other hand, if the interaction between the RTK and the PTP is defined by a negative feedback, the system will have a unique dynamical solution that maintains low levels of phosphorylation of the receptor (Fig 3C). Typical examples of phosphatases that could work in this way are the cytosolic phosphatases Shp1/2 that are activated by binding to phosphotyrosine residues via an SH2 domain (Barford & Neel, 1998; Keilhack et al, 1998).
The response of both systems (toggle switch and the negative feedback) to growth factor stimuli is thus uniquely determined by their respective topology. In case of the toggle switch coupled to an autocatalytic RTK activation, a small amount of the ligand will initially activate a small amount of receptors. This activated fraction however will suffice to push the system over the threshold and fully activate all other receptors (Reynolds et al, 2003). An irreversible situation is thereby generated in which all receptors remain active (Fig 3B). In the case of the negative feedback however, the system will tend to relax back to the original low phosphorylation state after stimulation and therefore lead to a pulse of receptor activity—an adaptive response (Fig 3C) (Tyson et al, 2003). Signal duration is therefore commonly regulated via negative feedback motifs.
The above examples show how intracellular network topology gives rise to differential dynamics in single cells, and even plasticity in signaling responses to growth factors. This description does not require the full details of the reactions that generate the communicating chemical intermediates, but only the directionality and type of interactions (Fig 2B and C). The relation between dynamics and causality (and vice versa) is however not a one‐to‐one mapping (Box 3). A given dynamical behavior can result from several different network topologies (Kirshna et al, 2009), but also a single network can display multiple stable dynamical solutions (Koseska et al, 2007). In the formulation of dynamical systems theory, these solutions are called attractors (Box 4) (Strogatz, 2001). Each attractor represents a distinct state of the system in phase space (Strogatz, 2001) that is stable within a given parameter range. This basin of stability reflects the robustness of the dynamical solution to external perturbations or stochastic fluctuations (Menck et al, 2013). Characteristic network topologies that display a rich phase space combine activatory and inhibitory interactions (Kirshna et al, 2009; Kaluza & Meyer‐Ortmanns, 2010). In particular, the ratio between the two types of causal interactions suggests whether the system can have multiple dynamical solutions (Kaluza & Meyer‐Ortmanns, 2010; Hong & Strogatz, 2011; Levnajić, 2012). Thus, cells use simple network motifs as building blocks to generate multiple attractors that enable flexibility in the responsiveness of the system. This implies that even genetically identical cells can respond differently to input signals because they are in different attractors, as dependent on initial conditions. For example, two completely identical cells that contain the RTK–PTP toggle switch discussed above can have two opposed states of the RTK: fully on or off (Fig 3B). Which of the two states is attained depends on the history of the cell as conditioned by previous stimuli. The same two states can be also observed in a population of genetically identical cells with different RTK–PTP expression levels due to the naturally occurring variance in protein abundances that results from gene expression noise. Thus, the multiple attractors that can arise from a fixed network configuration provide plasticity to the system.
Box 3: Network topology and dynamics.
The possible dynamical solutions of a given network are determined by the underlying topology (directed activatory or inhibitory links) and the dynamical behavior of the chemical intermediates that represent the nodes of the network. Let us consider a simple case of two interacting components: Protein X stimulates its own production (positive feedback) and that of protein Y, whereas Y inhibits X by transcriptional or translation regulation, or degradation. The positive feedback from X to itself generates bistable behavior that can be observed for different maximum rate production (Angeli et al, 2004; Mitrophanov & Groisman, 2008). Generating the negative feedback loop between X and Y on the other hand establishes the necessary preconditions for oscillations. For a particular dynamics of the nodes, for example, when the half‐life of the protein Y is smaller than that of X, oscillatory behavior will be observed (Kirshna et al, 2009).
How does the dynamics of this system evolve? In the absence of the negative feedback loop, namely Y is held fixed and the system is bistable for a certain range of Y values. Thus, depending on the initial conditions, the system will be either in high‐ or low‐activity states of protein X (dark blue lines). When the regulation from X to Y is established, the system displays oscillatory behavior: When the underlying bistability pushes X toward the high activity state, more Y is produced leading to a decrease in X, and vice versa for the low state, enabling the oscillations to continue (light blue lines).
Different causal relations however can give rise to the same dynamical behavior like bistability and oscillations as shown in (B). The degeneracy of the problem further increases with increased number of nodes in the network (C).
Box 4: Concepts from dynamical systems theory.
Consider a cellular state that is specified in terms of the concentrations of the active protein states. For example, if upon a given stimulus, k signaling proteins are activated, the cellular response, and thereby its state is described by the concentrations of those proteins (x(t), y(t), z(t), …) over time. One state of the system, that is, one combination of these proteins represents a point in vector space that is called phase space. The dynamics of the system is then described by the dynamics of the corresponding phase space points in terms of their trajectories. With time, the phase variables reach a stable range of values. This bounded region of phase space is called attractor and represents dynamical solution of the system that is invariant with time. The attractors can be fixed points representing stationary states, limit cycles (periodic oscillations), and strange attractors (chaotic behavior). Different dynamical solutions that a system can reach depending on the initial values of the variables are represented as different attractors in phase space. Staring from a given set of initial values, the system will tend to evolve toward a particular attractor. The regions of such initial values, called basins of attraction or basins of stability, partition the phase space around the stable attractors. The system can therefore jump between different attractors if large enough perturbations are applied.
An additional level of plasticity is realized when causal connections are conditioned by other ones. The conditioning can be stimulus dependent, enabling diverse signals to be processed through the same cascade. An example is the mitogen‐activated protein kinase (MAPK) cascade of Raf, Mek, and Erk (Marshall, 1995; Lewis et al, 1998), where the phosphorylated active forms of the kinases represent the communicating intermediates of the nodes (Kholodenko et al, 2002). The positive feedback between Erk and Raf in this case is conditioned on nerve growth factor (NGF)‐dependent protein kinase C (PKCδ) phosphorylation of Raf kinase inhibitor protein (RKIP) that removes it from Raf (Santos et al, 2007). In network formalism, this directed link is not between the two nodes RKIP and RAF, but rather the interaction between PKCδ and RKIP enables the causal interaction between Erk and Raf to take place (Fig 4). This results in a sustained Erk activity profile. On the other hand, epidermal growth factor (EGF) stimulation does not lead to PKCδ activation and only the negative feedback between Erk and Raf manifests. This now results in a transient Erk activity profile. This NGF/EGF stimulus‐dependent conditioning of the causal interactions thus leads to different MAPK dynamics that cause cellular differentiation and proliferation, respectively. Recently, it has been postulated that this negative feedback is also conditioned on the activity of the epidermal growth factor receptor, which might further discriminate the two input signals by the MAPK module (Ryu et al, 2015).
Understanding how a given dynamical behavior arises necessitates network reconstruction to identify the underlying topology of the network. This information then allows to investigate the potential of cells in terms of dynamical solutions and thereby their plasticity in responses.
Deriving causality from dynamics
It is exactly the dynamical behavior of the system's components, that is, the active states of signaling molecules as a function of time, that is the subject of experimental observation (Santos et al, 2007; Ryu et al, 2015). Importantly, the observed dynamical responses of the nodes do not reflect the kinetic properties of the molecules in isolation, but rather the dynamics of the network in which they are embedded. A given measurement reveals only one dynamical solution that the protein interaction network can exhibit, which is dependent on the initial conditions of the nodes. To reconstruct a causal network from this dynamics of its constituents, it is necessary to exploit the property that coupled events have temporal order. One possibility is to impose a causal relation of a “driver component” and a “response component” on the nodes.
This is accomplished by combining experimental perturbations with observations of the chemical intermediates. The perturbations need to be small, to maintain the system in the same dynamical solution. Quantification of the corresponding changes in the nodes in terms of global response coefficients allows reconstructing the underlying causal relationships by computing the local response coefficients using Modular Response Analysis (MRA, Box 5A) (Bruggeman et al, 2002; Santos et al, 2007; Klinger et al, 2013). In reality, the dynamics of only a limited set of nodes is observed within the context of the complete proteome. MRA allows uncovering the causal interactions between the measured nodes, as conditioned by the unobserved components of the proteome. It is therefore necessary to consider whether chronic genetic perturbations alter the context of the hidden network by affecting gene expression. Genetic perturbation should be thus ideally complemented by acute perturbations using, for example, small molecule inhibitors, pharmaco‐ or optogenetic perturbations that maintain the same proteome context (Zamir & Bastiaens, 2008; Toettcher et al, 2013; Kim et al, 2014).
Box 5: Principles of network reconstruction.
A. To reconstruct a causal network from the dynamics of its constituents, the property that coupled events have temporal order is exploited. Modular Response Analysis (MRA) uses this principle by imposing causality on the system via sequential node perturbations (Bruggeman et al, 2002). The corresponding changes in the remaining components of the system are thereby quantified as a means to infer directional relationships between the nodes in the network. One can thus measure the response of protein pi to the perturbation in protein pj (P), as mediated by the entire network after reaching a steady state. From this global response coefficients (Rij = ∂pi/∂Ρ) matrix, the local response coefficients rij = ∂pi/∂pj indicating the causal connectivity between pi and pj can be derived according to ∑r ik R kj = 0. rij > 0 indicates activatory links, rij < 0 inhibitory links, and rij = 0 no direct links between the nodes pi and pj.
B. Stepwise network inference from time‐resolved data can be also accomplished using association measures. At a distinct time point after stimulus, the extent of modification of some property of the nodes, that is, gene expression or protein phosphorylation, is measured for all nodes simultaneously. Experimental acquisition of this information at n (non)uniformly distributed time points thereby reflects the dynamics of each of the nodes in the network. Since the temporal profiles of two nodes that regulate each other are similar, the regulatory links can be derived by thresholding the pairwise similarities in the association matrix. Measures operating on vectors, such as classical distance measures and dynamic time warping, random variables (correlations, permutation‐based and information‐theoretic measures), model‐based measures (Granger causality) and measures operating on symbolic dynamics can be used to derive the association matrix (Hempel et al, 2011a). Applying additionally partial variants of the measures to account for indirect regulations and scoring rules for the pairwise weighting of the interactions, the number of falsely identified links can be reduced (summary on the methods and scoring schemes is presented in Hempel et al, 2011a). Since the majority of these methods and scoring schemes are symmetric, it is necessary to apply time shifting or symmetry breaking schemes to infer the causality of the identified links.
C. Bayesian networks, a form of graphical models, have been also used to reconstruct causal signaling networks from multivariate datasets, for example, the phosphorylation state of multiple proteins in a same cell. They explicitly make use of the naturally occurring cell‐to‐cell variance, since Bayesian networks can represent probabilistic non‐linear dependence relationships among multiple interacting components. The analysis algorithm therefore approximates the network topology that closely predicts the made observations by traversing the space of possibilities via single‐link changes that improve the score of the model (Sachs et al, 2005). Although the obtained relationships are statistical in nature, causality of the interactions could be obtained when experimental perturbations are additionally applied.
Applying MRA to reconstruct large‐scale networks is however experimentally challenging, since the number of combinations between perturbations and node observations scales quadratic with the number of nodes (Box 5A). Therefore, other methods that uncover the asymmetry of causal interactions without necessarily using perturbations are required to deduce topology. This can be achieved by applying asymmetric measures to time‐resolved biochemical reaction data. In these approaches, the similarity between the activity profiles is quantified, while implicitly or explicitly accounting for time delays in the responses of the nodes (Box 5B). Similarly to the reconstruction of gene regulatory networks (Wang et al, 2008; Hempel et al, 2011a,b; Ma et al, 2014), it is possible to apply these methods to time‐resolved proteomics data to identify the flow of information in the system. Currently, all of the time series analysis methods have a limited accuracy in the number of identified links (~80/30% true/false positives). This strongly depends on the size of the network and the temporal resolution of the measured profiles (Hempel et al, 2011b). To compensate for the limited information contained in sparsely sampled time series, the activity states of several proteins have been simultaneously measured in single cells (Sachs et al, 2005; Kirshnaswamy et al, 2014). By having hundred individual cells, each providing a point of multidimensional data, cell‐to‐cell variability has been used to infer and quantify (non)linear relationships between proteins using different statistical approaches. For example, Bayesian analysis (Box 5C) has been applied to reconstruct protein interaction networks from primary human immune system single‐cell data obtained by flow cytometry (Sachs et al, 2005). The cell‐to‐cell variability in the measured activity state of multiple phosphorylated proteins and phospholipid components as obtained by node perturbations has been incorporated in a Bayesian model to derive probabilistic dependences among multiple components. In another study, conditional density‐based analysis of single‐cell mass cytometry data has been developed to identify and quantify the functional dependencies between proteins that participate in antigen recognition in cells of the immune system (Kirshnaswamy et al, 2014). The next challenge will be to combine the associations between multiple network components obtained at distinct time points in a single measure. This would provide a theoretical framework that combines the information contained in the cell‐to‐cell variance with the information that the temporal profiles encode, resulting most likely in more efficient methods for reconstruction of causal protein interaction networks.
Recursive communication between cells leads to cognition
In the previous examples of signaling network reconstruction, external growth factors were imposed, thereby enforcing a unidirectional information flow from the environment to the inside of the cell. However, the collective behavior of cells that leads to coordinated dynamical behavior, such as the behavior of cells in an organ or a tissue (Perbal, 2003; Harper et al, 2010), requires constant adjustment of the internal state by not only signal reception, but also emission of information about the internal state to neighboring cells. This iterative exchange of information that occurs via cell‐cell communication can therefore give rise to cooperative cellular behavior even under noisy conditions (Koseska et al, 2009; Box 6). For example, most physiological events in mammals are subjected to well‐controlled daily oscillations that are generated by an internal self‐sustained circadian oscillator located in individual neurons in the hypothalamic suprachiasmatic nucleus (SCN) (Welsh et al, 1995; Moore, 1997). In this case, a coherent and synchronous circadian output is produced as a result of intercellular recursive communication via neurotransmitters (Yamaguchi et al, 2003).
Box 6: Stabilizing cellular responses by intercellular communication.
Cooperative cellular responses are generally subject to fluctuations in intercellular signals that are themselves processed by biochemical networks in which the level of the components is also subject to stochasticity. In addition to the observations that transcriptional noise can be filtered out by cell‐intrinsic factors, that is, given topology of gene regulatory networks (Thattai & van Oudenaarden, 2000; Hooshangi et al, 2004), it has been also hypothesized that intercellular communication can enhance the reliability of cellular responses in noisy conditions (Koseska et al, 2009). In particular, it has been shown that cells reach a particular fate only when a specific size of the population is reached.
A theoretical basis for this has been established in general non‐linear stochastic models, where noise is known to be tunable through the size of the system such that the amplitude of the noise is inversely proportional to the square root of number of cells (Pikovsky et al, 2002).
Even more, intercellular recursive communication can give rise to new attractors different than those of the isolated cells (Koseska et al, 2007; Ullner et al, 2008a,b), thereby displaying cognitive behavior. Analogous are the observations that individual hematopoietic stem cells acquire tissue‐like properties as a result of microenvironmental cues from the liver (Jang et al, 2004). Interestingly, specific communication‐induced dynamical solutions allow for heterogeneity to be established and maintained in a population (Koseska et al, 2010), resembling differentiated cellular entities in tissues. Theoretical studies have suggested that novel dynamical solutions can be generated in systems characterized with bidirectional inhibitory intercellular communication, rendering cognition a generic property of such multicellular systems (Koseska et al, 2007; Ullner et al, 2008a,b).
To demonstrate this idea, let us consider again the RTK–PTP toggle switch example described above. This network can exhibit one of two possible stable steady states in isolated cells, dependent on prior exposure to growth factor. This is a result of bistability that is generated due to the RTK‐induced PTP inhibition via ROS (Fig 5A). However, when ROS diffuses across the membrane, the short‐range bidirectional coupling between the networks in neighboring cells can theoretically generate a novel dynamical solution—an inhomogeneous steady state (IHSS, Fig 5B). This solution occurs due to symmetry breaking of the steady state via a pitchfork bifurcation and is characterized with two stable branches. Thus, even when the two communicating networks of the neighboring cells start from identical initial conditions, one of them must adopt the high branch, whereas the other cell must adopt the low branch. This will result in one cell having high and the other a low level of phosphorylated RTK. This IHSS solution coexists in parameter space with the bistability region (Fig 5B), but is a distinct dynamical solution of the coupled networks. Moreover, the IHSS (Koseska et al, 2007) is a generalized Turing instability (Turing, 1952; Challenger et al, 2015) that occurs via a communication‐induced Turing‐type bifurcation (Koseska et al, 2013). This solution has been theoretically linked to emergence of heterogeneous dynamical behavior in a homogeneous multicellular population as a way to establish asymmetric cell differentiation (Koseska et al, 2010). The IHSS solution has also been related to striped pattern formation that results from Delta–Notch signaling (Fig 1). In this case, the IHSS is driven by a primary signaling source and the addition of cis‐interactions (Formosa‐Jordan & Ibanes, 2014). Expanding the size of the population can further increase the number of dynamical solutions and thereby the differentiation potential in the system by means of clustering distinct manifestations of the same dynamical solution (Koseska et al, 2010).
The described principles are not specific for the RTK–PTP interaction network considered here, but is rather an inherent property of coupled systems, since it relies on the dynamics of the underlying networks as determined by their topology. For example, single cells that contain a three‐component genetic repressilator network motif (Elowitz & Leibler, 2000) can exhibit a single dynamical solution—autonomous oscillations with a defined period (Fig 6). Coupling these networks via a small signaling molecule however generates not only an IHSS, but also an inhomogeneous limit cycle solution, IHLC (Fig 6, Ullner et al, 2008b). This IHLC mimics the cellular potential to both proliferate and differentiate, and has been therefore related to asymmetric stem cell differentiation with self‐renewal (Suzuki et al, 2011).
Thus, paracrine intercellular communication can generally result in collective dynamic behavior that is different from that of isolated cells, which might account for tissue‐specific cell functionalities. The above‐described hypotheses therefore state that cell identity is not purely determined by the manifestation of the genome but rather defined by recursive intercellular communication. The inherent multistability of multicellular systems defines the range in which cells can respond and adapt, while still maintaining their identity. In this description, stable attractors are associated with specific functional outputs, giving a mathematical formalism to cellular information processing. A further extension of the idea is the possibility for cells to modulate their dynamics on the proteome level and thereby acquire new functions. Making the parallel to neural networks and the concept of reservoir computing (Box 7), it is theoretically possible for recurrent networks that employ dynamic processes on different timescales to process information using transiently stable attractors (Maass et al, 2002). This significantly increases the network capabilities for information processing, especially of time‐varying inputs that can be further translated to novel cellular functions. If it can be realized on the level of the proteome, this could imply that the identity of cells can be altered by “teaching” them how to perceive the environment.
Box 7: Reservoir computing.
Reservoir computing is a machine‐learning concept referring to models that rely on mapping of stimuli on a high‐dimensional space in a non‐linear fashion. Such models include echo‐state and liquid‐state machines and state‐dependent networks (Buonomano & Maass, 2009). This concept for real‐time processing of time‐varying inputs requires recurrent networks whose components interact with each other via diverse processes with different timescales and transfer functions, for example, phosphorylation, enzymatic degradation, transcriptional, post‐transcriptional, and post‐translational regulation. In this view, each node in the network receives time‐varying input from external sources as well as from other nodes. The recurrent nature of the connections turns the time‐varying input into spatial–temporal patterns of activation in the network nodes, which are read out by linear discriminant units.
What determines the identity of cancer cells?
The collective behavior of cells on the level of tissues is generally stable against mutations that occur in single cells, allowing them to retain their identity (Bissel & Hines, 2011). The question therefore is how the cancer phenotype is stabilized under these conditions. When an oncogenic “gain‐of‐function” mutation occurs in single cells, it constitutively activates a certain protein. In terms of the intracellular signaling network, the continuous activity of this node “pushes” the complete network to a particular dynamical state. If we assume that the topology is not affected by the oncogene expression, then the number of attractors in these cells is reduced. However, comparative bioinformatics analysis of microarray datasets from multiple tumors and the non‐malignant tissues they were derived from has identified significantly increased number of interactions and multi‐edge node pairs in the regulatory networks of tumor cells (Schramm et al, 2010; Rahman et al, 2013). The formation of these new connections likely results from changes in gene expression that lead to an altered proteome composition (Schramm et al, 2010). Another possibility is that oncogene overexpression leads to new interactions with effectors by a simple mass action effect (Jones et al, 2006). Thus, a significant rewiring of the signaling networks in cells where oncogenic mutations occur will effectively modulate their dynamical possibilities by increasing the number of attractors. This would lead to an increased plasticity of cancer cells as compared to the tissue cells they are derived from. That this might be the case is evident from the fact that some cancer cells reversibly transit among states that differ in their competence, as a means to explore possibilities to contribute to tumor growth (Mani et al, 2008; Gupta et al, 2011).
This also allows cells to “jump” to a new “selfish” attractor that is intrinsically stabilized in single cells (Kaneko, 2011). Due to the acquired changes in dynamics, these cells loosen their collective behavior in the tissue and start to behave more as single entities. Since they represent an aberrant cell type, they are functionally unnecessary for the organism and therefore have not achieved robustness against noise and mutation during evolution (Kaneko, 2011). This leaves the dynamical state of these cells more changeable to somatic mutations, allowing them to continuously transit between different attractors (Capp, 2005). Selection pressure would eventually drive them in a state with a larger basin of attraction, giving rise to a stable cancer cell characterized by increased signaling entropy (Teschendorff et al, 2014; Box 8). The incidence of intercellular communication between cancer cells is now higher than that to the normal tissue, thereby stabilizing the tumor identity (Hamada et al, 1988). To which extent the dynamics of communicating signaling networks can be trained by environmental cues to maintain a specific behavior in a tissue context can be thus applied both, to understand how cancer cells acquire metastatic behavior, but also whether cancer cells can be “retrained” by the surrounding tissue to adapt their behavior. It has been demonstrated, for example, that transplantation of cancer cells into other tissues in which their interaction with other cells is modified, led to “reprogramming” of their oncogenic behavior (Mintz & Illmensee, 1975; Hochedlinger et al, 2004; Kasemeier‐Kulesa et al, 2008).
Box 8: Redundancy of signaling routes and signaling entropy.
The highly interconnected networks in transformed cells increase the redundancy in signaling paths that can be quantified through the concept of signaling entropy (West et al, 2012). Increasing the number of routes through which information can flow increases the signaling entropy. The high signaling entropy of cancer cells makes them robust to inhibition of protein interactions (Teschendorff et al, 2014), implying that pharmacological targeting of an oncogene product alone does not necessarily affect cell transformation. The topology of the transformed network, especially the positioning of negative feedbacks that can lead to homeostatic behavior against pharmacological perturbations, plays thereby a crucial role. For example, a negative feedback from Erk to EGFR has been shown to lead to AKT cross‐activation upon MEK inhibition in cells with oncogenic KRas or BRaf. A successful therapy for colon cancer therefore requires combined inhibition of MEK and EGFR to prevent AKT activation and tumor cell survival (Klinger et al, 2013).
Conclusion
Signaling cannot be perceived as a unidirectional process from the outside to the inside of the cell that is executed by hardwired cellular machinery. It is rather a dynamical manifestation of a highly adaptive network that is determined by the recursive interactions between the protein reaction states within the network as well as with the networks of other cells with which it communicates by extracellular chemical means. In this way, the processing of information becomes dependent on historical and extracellular context. We therefore like to think about signaling in cellular ensembles as a process reminiscent to cognition. In order to understand how this cognitive process works, inferring the topology of the network of signaling networks is a prerequisite to relate the observed biochemical behavior to the general principles of information processing between cells, and the thereby established cellular function. It is thus important to consider that new dynamical solutions can arise from intercellular communication and that cognition is not a property of single cells per se. This in turn opens the possibilities to manipulate cellular identity by changing its interaction with the environment: the multicellular context in which it is embedded.
Conflict of interest
The authors declare that they have no conflict of interest.
Supporting information
The EMBO Journal (2017) 36: 568–582
Contributor Information
Aneta Koseska, Email: aneta.koseska@mpi-dortmund.mpg.de.
Philippe IH Bastiaens, Email: philippe.bastiaens@mpi-dortmund.mpg.de.
References
- Angeli D, Ferrell JE Jr, Sontag ED (2004) Detection of multistability, bifurcations, and hysteresis in a large class of biological positive‐feedback systems. Proc Natl Acad Sci USA 101: 1822–1827 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barford D, Neel BG (1998) Revealing mechanisms for SH2 domain mediated regulation of the protein tyrosine phosphatase SHP‐2. Curr Biol 6: 249–254 [DOI] [PubMed] [Google Scholar]
- Bissel MJ, Hines WC (2011) Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat Med 17: 320–329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bourgine P, Stewart J (2004) Autopoiesis and cognition. Artif Life 10: 327–345 [DOI] [PubMed] [Google Scholar]
- Brandman O, Ferrell JE Jr, Rong L, Meyer T (2005) Interlinked fast and slow positive feedback loops drive reliable cell decisions. Science 310: 496–498 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bruggeman FJ, Westerhoff HV, Hoek JB, Kholodenko BN (2002) Modular response analysis of cellular regulatory networks. J Theor Biol 218: 507–520 [PubMed] [Google Scholar]
- Buonomano DV, Maass W (2009) State‐dependent computations: spatiotemporal processing in cortical networks. Nat Rev Neurosci 10: 113–125 [DOI] [PubMed] [Google Scholar]
- Capp JP (2005) Stochastic gene expression, disruption of tissue averaging effects and cancer as a disease of development. Bioessays 27: 1277–1285 [DOI] [PubMed] [Google Scholar]
- Challenger JD, Burioni R, Fanelli D (2015) Turing‐like instabilities from limit cycle. Phys Rev E Stat Nonlin Soft Matter Phys 92: 022818 [DOI] [PubMed] [Google Scholar]
- Cox J, Mann M (2011) Quantitative, high‐resolution proteomics for data‐driven systems biology. Annu Rev Biochem 80: 273–299 [DOI] [PubMed] [Google Scholar]
- Denu JM, Tanner KG (1998) Specific and reversible inactivation of protein tyrosine phosphatases by hydrogen peroxide: evidence for a sulfenic acid intermediate and implications for redox regulation. Biochemistry 37: 5633–5642 [DOI] [PubMed] [Google Scholar]
- D'Souza RCJ, Knittle AM, Nagaraj N, van Dinther M, Choudhary C, ten Dijke P, Mann M, Sharma K (2014) Time‐resolved detection of early phosphoproteome and ensuing proteome changes in response to TGF‐beta. Sci Signal 7: rs5 [DOI] [PubMed] [Google Scholar]
- Elowitz M, Leibler S (2000) A synthetic oscillatory network of transcriptional regulators. Nature 403: 335–338 [DOI] [PubMed] [Google Scholar]
- Farnsworth KD, Nelson J, Gershenson C (2013) Living is information processing: from molecules to global systems. Acta Biotheor 61: 203–222 [DOI] [PubMed] [Google Scholar]
- Finkel T (1998) Oxygen radicals and signaling. Curr Opin Cell Biol 10: 248–253 [DOI] [PubMed] [Google Scholar]
- Fong SS, Palsson BO (2004) Metabolic gene deletion strains of E. coli evolve to computationally predicted growth phenotypes. Nat Genet 36: 1056–1058 [DOI] [PubMed] [Google Scholar]
- Formosa‐Jordan P, Ibanes M (2014) Competition in Notch signaling with cis enriches cell fate decisions. PLoS One 9: e95744 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Francavilla C, Hekmat O, Blagoev B, Olsen J (2014) SILAC‐based temporal phosphoproteomics. Methods Mol Biol 1188: 125–148 [DOI] [PubMed] [Google Scholar]
- Gavin AC, Bösche M, Krause R, Grandi P, Marzioch M, Bauer A, Schultz J, Rick J, Michon AM, Cruciat CM, Remor M, Höfer C, Schelder M, Brajenovic M, Ruffner H, Merino A, Klein K, Hudak M, Dickson D, Rudi T et al (2002) Functional organization of the yeast proteome by systematic analysis of protein complexes. Nature 415: 141–147 [DOI] [PubMed] [Google Scholar]
- Grecco HE, Schmick M, Bastiaens PIH (2011) Signaling from the living plasma membrane. Cell 144: 897–909 [DOI] [PubMed] [Google Scholar]
- Gupta PB, Fillmore CM, Jiang G, Shapira SD, Tao K, Kuperwasser C, Lander ES (2011) Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 146: 633–644 [DOI] [PubMed] [Google Scholar]
- Hamada J, Takeichi N, Kobayashi H (1988) Metastatic capacity and intercellular communication between normal cells and metastatic cell clones derived from a rat mammary carcinoma. Cancer Res 48: 5129–5132 [PubMed] [Google Scholar]
- Harper CV, Featherstone K, Semprini S, Friedrichsen S, McNeilly J, Paszek P, Spiller DG, McNeilly AS, Mullins JJ, Davis JRE, White MRH (2010) Dynamic organization of prolactin gene expression in living pituitary tissue. J Cell Sci 123: 424–430 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hempel S, Koseska A, Kurths J, Nikoloski Z (2011b) Inner composition alignment for inferring directed networks from short time series. Phys Rev Lett 107: 054101 [DOI] [PubMed] [Google Scholar]
- Hempel S, Koseska A, Nikoloski Z, Kurths J (2011a) Unraveling gene regulatory networks from time‐resolved gene expression data – a measures comparison study. BMC Bioinformatics 12: 292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hochedlinger K, Blelloch R, Brennan C, Yamada Y, Kim M, Chin L, Jaenisch R (2004) Reprogramming of a melanoma genome by nuclear transplantation. Genes Dev 18: 1875–1885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hong H, Strogatz S (2011) Kuramoto model of coupled oscillators with positive and negative coupling parameters: an example of conformist and contrarian oscillators. Phys Rev Lett 106: 054102 [DOI] [PubMed] [Google Scholar]
- Hooshangi S, Thiberge S, Weiss R (2004) Ultrasensitivity and noise propagation in a synthetic transcriptional cascade. Proc Natl Acad Sci USA 102: 3581–3586 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Humphrey SJ, Azimifar SB, Mann M (2015) High‐throughput phosphoproteomics reveals in vivo insulin signaling dynamics. Nat Biotechnol 33: 990–995 [DOI] [PubMed] [Google Scholar]
- Ibarra RU, Edwards JS, Palsson BO (2002) E. coli K‐12 undergoes adaptive evolution to achieve in silico predicted optimal growth. Nature 420: 186–189 [DOI] [PubMed] [Google Scholar]
- Jacob F (1977) Evolution and tinkering. Science 196: 1161–1166 [DOI] [PubMed] [Google Scholar]
- Jang Y, Collector MI, Baylin SB, Diehl AM, Sharkis SJ (2004) Hematopoetic stem cells convert into liver cells within days without fusion. Nat Cell Biol 6: 532–539 [DOI] [PubMed] [Google Scholar]
- Jones RB, Gordus A, Krall JA, Macbeath G (2006) A quantitative protein interaction network for ErbB receptors using protein microarrays. Nature 439: 168–174 [DOI] [PubMed] [Google Scholar]
- Kaluza P, Meyer‐Ortmanns H (2010) On the role of frustration in excitable systems. Chaos 20: 043111 [DOI] [PubMed] [Google Scholar]
- Kaneko K (2011) Characterization of stem cells and cancer cells on the basis of gene expression profile stability, plasticity, and robustness. Bioessays 33: 403–413 [DOI] [PubMed] [Google Scholar]
- Kasemeier‐Kulesa JC, Teddy JM, Postovit LM, Seftor EA, Hendrix MJ, Kulesa PM (2008) Reprogramming multipotent tumor cells with the embryonic neural crest microenvironment. Dev Dyn 237: 2657–2666 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kashiwagi A, Urabe I, Kaneko K, Yomo T (2006) Adaptive response of a gene network to environmental changes by fitness‐induced attractor selection. PLoS One 1: e49 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keilhack H, Tenev T, Nyakatura E, Godovac‐Zimmerman J, Nielsen L, Seedorf K, Bohmer FD (1998) Phosphotyrosine 1173 mediates binding of the protein tyrosine phosphatase SHP‐1 to the epidermal growth factor receptor and attenuation of receptor signaling. J Biol Chem 73: 24839–24846 [DOI] [PubMed] [Google Scholar]
- Kholodenko BN, Kiyatkin A, Bruggeman FJ, Sontag E, Westerhoff HV, Hoek J (2002) Untangling the wires: a strategy to trace functional interactions in signaling and gene networks. Proc Natl Acad Sci USA 99: 12841–12846 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kiel C, Verschueren E, Yang J, Serrano L (2013) Integration of protein abundance and structure data reveals competition in the ErbB signaling network. Sci Signal 6: ra109 [DOI] [PubMed] [Google Scholar]
- Kim N, Kim JM, Lee M, Kim CY, Chang K, Heo WD (2014) Spatiotemporal control of Fibroblast growth factor receptor signals by blue light. Chem Biol 21: 903–912 [DOI] [PubMed] [Google Scholar]
- Kirshna S, Semsey S, Jensen MH (2009) Frustrated bistability as a means to engineer oscillations in biological systems. Phys Biol 6: 036009 [DOI] [PubMed] [Google Scholar]
- Kirshnaswamy S, Spitzer MH, Mingueneau M, Bendall SC, Litvin O, Stone E, Pe'er D, Nolan G (2014) Conditional density‐based analysis of T cell signaling in single‐cell data. Science 346: 1250689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klinger B, Sieber A, Fritsche‐Guenther R, Witzel F, Berry L, Schumacher D, Yan Y, Durek P, Merchant M, Schäfer R, Sers C, Blüthgen N (2013) Network quantification of EGFR signaling unveils potential for targeted combination therapy. Mol Syst Biol 9: 673 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koseska A, Volkov E, Zaikin A, Kurths J (2007) Inherent multistability in arrays of autoinducer coupled genetic oscillators. Phys Rev E Stat Nonlin Soft Matter Phys 75: 031916 [DOI] [PubMed] [Google Scholar]
- Koseska A, Zaikin A, Kurths J, Garcia‐Ojalvo J (2009) Timing cellular decision making under noise via cell‐cell communication. PLoS One 4: e4872 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koseska A, Ullner E, Volkov E, Kurths J, Garcia‐Ojalvo J (2010) Cooperative differentiation through clustering in multicellular populations. J Theor Biol 263: 189–202 [DOI] [PubMed] [Google Scholar]
- Koseska A, Volkov E, Kurths J (2013) Transition from amplitude to oscillation death via Turing bifurcation. Phys Rev Lett 111: 024103 [DOI] [PubMed] [Google Scholar]
- Larance M, Lamond AI (2015) Multidimensional proteomics for cell biology. Nat Rev Mol Cell Biol 16: 269–280 [DOI] [PubMed] [Google Scholar]
- Lemmon MA, Schlessinger J (2010) Cell signaling by receptor tyrosine kinases. Cell 141: 1117–1134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levnajić Z (2012) Evolutionary design of non‐frustrated networks of phase‐repulsive oscillators. Sci Rep 2: 967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lewis TS, Shapiro PS, Ahn NG (1998) Signal transduction through MAP kinase cascades. Adv Cancer Res 74: 49–139 [DOI] [PubMed] [Google Scholar]
- Ma H, Aihara K, Chen L (2014) Detecting causality from nonlinear dynamics with short‐term time series. Sci Rep 4: 7464 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maass W, Natschläger T, Markram H (2002) Real‐time computing without stable states: a new framework for neural computation based on perturbation. Neural Comput 14: 2531–2560 [DOI] [PubMed] [Google Scholar]
- Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA (2008) The epithelial‐mesenchymal transition generates cells with properties of stem cells. Cell 133: 704–715 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marshall CJ (1995) Specificity of receptor tyrosine kinase signaling: transient versus sustained extracellular signal‐regulated kinase activation. Cell 80: 179–185 [DOI] [PubMed] [Google Scholar]
- Maturana HR, Varela FJ (1980) Autopoiesis and cognition: the realization of the living. London, UK: D. Reidel Publishing Company; [Google Scholar]
- Menck PJ, Heitzig J, Marwan N, Kurths J (2013) How basin stability complements the linear‐stability paradigm. Nat Phys 9: 89–92 [Google Scholar]
- Meng TC, Fukada T, Tonks NK (2002) Reversible oxidation and inactivation of protein tyrosine phosphatases in vivo . Mol Cell 9: 387–399 [DOI] [PubMed] [Google Scholar]
- Mintz B, Illmensee K (1975) Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc Natl Acad Sci USA 72: 3585–3589 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitrophanov AY, Groisman EA (2008) Positive feedback in cellular control systems. Bioessays 30: 542–555 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore RY (1997) Circadian rhythms: basic neurobiology and clinical applications. Annu Rev Med 48: 253–266 [DOI] [PubMed] [Google Scholar]
- Ostman A, Bohmer D (2001) Regulation of receptor tyrosine kinase signaling by protein tyrosine phosphatases. Trends Cell Biol 11: 258–266 [DOI] [PubMed] [Google Scholar]
- Perbal B (2003) Communication is the key. Cell Commun Signal 1: 1–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pikovsky A, Zaikin A, de la Casa M (2002) System size resonance in coupled noisy systems and in the Ising model. Phys Rev Lett 88: 050601 [DOI] [PubMed] [Google Scholar]
- Prigogine I, Nicolis G (1967) On symmetry‐breaking instabilities in dissipative systems. J Chem Phys 46: 3542–3550 [Google Scholar]
- Rahman KMT, Islam F, Banik RS, Honi U, Diba FS, Sumi SS, Kabir ST, Akhter S (2013) Changes in protein interaction networks between normal and cancer conditions: total chaos or ordered disorder? Netw Biol 3: 15–28 [Google Scholar]
- Reynolds AR, Tischer C, Verveer PJ, Rocks O, Bastiaens PIH (2003) EGFR activation coupled to inhibition of tyrosine phosphatases causes lateral signal propagation. Nat Cell Biol 5: 447–453 [DOI] [PubMed] [Google Scholar]
- Ryu H, Chung M, Dobrzynski M, Fey D, Blum Y, Lee SS, Peter M, Kholodenko BN, Jeon NL, Pertz O (2015) Frequency modulation of Erk activation dynamics rewires cell fate. Mol Syst Biol 11: 838–852 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sachs K, Perez O, Pe'er D, Lauffenburger DA, Nolan GP (2005) Causal protein‐signaling networks derived from multiparametric single‐cell data. Science 308: 523–529 [DOI] [PubMed] [Google Scholar]
- Santos SDM, Verveer PJ, Bastiaens PIH (2007) Growth factor‐induced MAPK network topology shapes Erk response determining PC‐12 cell fate. Nat Cell Biol 9: 324–330 [DOI] [PubMed] [Google Scholar]
- Schramm G, Kannabiran N, König R (2010) Regulation patterns in signaling networks of cancer. BMC Syst Biol 4: 162 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schrödinger E (1944) What is life? UK: Cambridge University Press; [Google Scholar]
- Stewart J (1993) Cognition without neurons: adaptation, learning and memory in the immune system. Cognitiva 5: 187–202 [Google Scholar]
- Stolovicki E, Dror T, Brenner N, Braun E (2006) Synthetic gene recruitment reveals adaptive reprogramming of gene regulation in yeast. Genetics 173: 75–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strogatz S (2001) Nonlinear dynamics and chaos: with applications to physics, biology, chemistry and engineering. Boulder, CO: Westview Press; [Google Scholar]
- Suzuki N, Furusawa C, Kaneko K (2011) Oscillatory protein expression dynamics endows stem cells with robust differentiation potential. PLoS One 6: e27232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teschendorff AE, Sollich P, Kuehn R (2014) Signaling entropy: a novel network‐theoretical framework for systems analysis and interpretation of functional omic data. Methods 67: 282–293 [DOI] [PubMed] [Google Scholar]
- Thattai M, van Oudenaarden A (2000) Intrinsic noise in gene regulatory networks. Proc Natl Acad Sci USA 98: 8614–8619 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toettcher JE, Weiner OD, Lim WA (2013) Using optogenetics to interrogate the dynamic control of signal transmission by the Ras/Erk module. Cell 155: 1422–1434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turing A (1952) The chemical basis of morphogenesis. Philos Trans R Soc Lond B Biol Sci 237: 37–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 [DOI] [PubMed] [Google Scholar]
- Ullner E, Koseska A, Kurths J, Volkov E, Kantz H, Garcia‐Ojalvo J (2008b) Multistability of synthetic genetic networks with repressive cell‐to‐cell communication. Phys Rev E Stat Nonlin Soft Matter Phys 78: 031904 [DOI] [PubMed] [Google Scholar]
- Ullner E, Zaikin A, Volkov E, Garcia‐Ojalvo J (2008a) Multistability and clustering in a population of synthetic genetic oscillators via phase‐repulsive cell‐to‐cell communication. Phys Rev Lett 99: 148103 [DOI] [PubMed] [Google Scholar]
- Völkel P, Le Faou P, Angrand PO (2010) Interaction proteomics: characterization of protein complexes using tandem affinity purification‐mass spectrometry. Biochem Soc Trans 38: 883–887 [DOI] [PubMed] [Google Scholar]
- Von Kriegsheim A, Baiocchi D, Birtwistle M, Sumpton D, Bienvenuti W, Morrice N, Yamada K, Lamond A, Kalna G, Orton R, Gilbert D, Kolch W (2009) Cell fate decisions are specified by the dynamic ERK interactome. Nat Cell Biol 11: 1458–1464 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, Wu M, Li Z, Chan C (2008) Short time‐series microarray analysis: methods and challenges. BMC Syst Biol 2: 58 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Welsh DK, Logothetis DE, Meister M, Reppert SM (1995) Individual neurons disassociated from rat suprachiasmatic nucleus express independently phased circadian firing rhythms. Neuron 14: 697–706 [DOI] [PubMed] [Google Scholar]
- West J, Bianconi G, Severini S, Teschendorff AE (2012) Differential network entropy reveals cancer system hallmarks. Sci Rep 2: 802 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamaguchi S, Isejima H, Matsuo T, Okura R, Yagita K, Kobayashi M, Okamura H (2003) Synchronization of cellular clocks in the suprachiasmatic nucleus. Science 302: 1408–1412 [DOI] [PubMed] [Google Scholar]
- Zamir E, Bastiaens PI (2008) Reverse engineering intracellular biochemical networks. Nat Chem Biol 4: 643–647 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.