scispace - formally typeset
Search or ask a question

Showing papers in "arXiv: Molecular Networks in 2010"


Posted Content
TL;DR: This article showed that regardless of the number of reactions in a metabolic genotype, the genotypes of a given phenotype typically form vast, connected, and unstructured sets that nearly span the whole of genotype space.
Abstract: Background: A metabolic genotype comprises all chemical reactions an organism can catalyze via enzymes encoded in its genome. A genotype is viable in a given environment if it is capable of producing all biomass components the organism needs to survive and reproduce. Previous work has focused on the properties of individual genotypes while little is known about how genome-scale metabolic networks with a given function can vary in their reaction content. Results: We here characterize spaces of such genotypes. Specifically, we study metabolic genotypes whose phenotype is viability in minimal chemical environments that differ in their sole carbon sources. We show that regardless of the number of reactions in a metabolic genotype, the genotypes of a given phenotype typically form vast, connected, and unstructured sets -- genotype networks -- that nearly span the whole of genotype space. The robustness of metabolic phenotypes to random reaction removal in such spaces has a narrow distribution with a high mean. Different carbon sources differ in the number of metabolic genotypes in their genotype network; this number decreases as a genotype is required to be viable on increasing numbers of carbon sources, but much less than if metabolic reactions were used independently across different chemical environments. Conclusions: Our work shows that phenotype-preserving genotype networks have generic organizational properties and that these properties are insensitive to the number of reactions in metabolic genotypes.

60 citations


Book ChapterDOI
TL;DR: Instead of modeling regulatory networks in terms of the deterministic dynamics of concentrations, this work model the dynamics of the probability of a given copy number of the reactants in single cells.
Abstract: The past decade has seen a revived interest in the unavoidable or intrinsic noise in biochemical and genetic networks arising from the finite copy number of the participating species. That is, rather than modeling regulatory networks in terms of the deterministic dynamics of concentrations, we model the dynamics of the probability of a given copy number of the reactants in single cells. Most of the modeling activity of the last decade has centered on stochastic simulation of individual realizations, i.e., Monte-Carlo methods for generating stochastic time series. Here we review the mathematical description in terms of probability distributions, introducing the relevant derivations and illustrating several cases for which analytic progress can be made either instead of or before turning to numerical computation.

54 citations


Journal ArticleDOI
TL;DR: In this article, a transfer matrix method was used to rigorously follow the system through the disruptive process of cell division, and the combined treatment of noisy dynamics both between and during cell division provided an efficient way to calculate the stability of alternative states in a broad range of epigenetic systems.
Abstract: Cells can often choose among several stably heritable phenotypes. Examples are the expression of genes in eukaryotic cells where long chromosomal regions can adopt persistent and heritable silenced or active states, that may be associated with positive feedback in dynamic modification of nucleosomes. We generalize this mechanism in terms of bistability associated with valleys in an epigenetic landscape. A transfer matrix method was used to rigorously follow the system through the disruptive process of cell division. This combined treatment of noisy dynamics both between and during cell division provides an efficient way to calculate the stability of alternative states in a broad range of epigenetic systems.

49 citations


Posted Content
TL;DR: The hypothesis that certain types of metabolic cycles may have played a role in the development of primitive metabolism despite the absence of regulatory mechanisms is supported.
Abstract: We investigate the stability properties of two different classes of metabolic cycles using a combination of analytical and computational methods. Using principles from structural kinetic modeling (SKM), we show that the stability of metabolic networks with certain structural regularities can be studied using a combination of analytical and computational techniques. We then apply these techniques to a class of single input, single output metabolic cycles, and find that the cycles are stable under all conditions tested. Next, we extend our analysis to a small autocatalytic cycle, and determine parameter regimes within which the cycle is very likely to be stable. We demonstrate that analytical methods can be used to understand the relationship between kinetic parameters and stability, and that results from these analytical methods can be confirmed with computational experiments. In addition, our results suggest that elevated metabolite concentrations and certain crucial saturation parameters can strongly affect the stability of the entire metabolic cycle. We discuss our results in light of the possibility that evolutionary forces may select for metabolic network topologies with a high intrinsic probability of being stable. Furthermore, our conclusions support the hypothesis that certain types of metabolic cycles may have played a role in the development of primitive metabolism despite the absence of regulatory mechanisms.

39 citations


Posted Content
TL;DR: The structure of the graphs with the smallest average distance and the largest average clustering given their order and size is described, and a method to make these networks more robust with respect to vertex removal is devised.
Abstract: We describe the structure of the graphs with the smallest average distance and the largest average clustering given their order and size. There is usually a unique graph with the largest average clustering, which at the same time has the smallest possible average distance. In contrast, there are many graphs with the same minimum average distance, ignoring their average clustering. The form of these graphs is shown with analytical arguments. Finally, we measure the sensitivity to rewiring of this architecture with respect to the clustering coefficient, and we devise a method to make these networks more robust with respect to vertex removal.

35 citations


Posted Content
TL;DR: It is demonstrated that the maximum genotype distance and robustness of metabolic networks can be explained by the number of superessential reactions and by the sizes of minimal metabolic networks viable in an environment.
Abstract: Metabolic networks are complex systems that comprise hundreds of chemical reactions which synthesize biomass molecules from chemicals in an organism's environment. The metabolic network of any one organism is encoded by a metabolic genotype, defined by a set of enzyme-coding genes whose products catalyze the network's reactions. Each metabolic genotype has a metabolic phenotype, such as the ability to synthesize biomass on a spectrum of different sources of chemical elements and energy. We here focus on sulfur metabolism, which is attractive to study the evolution of metabolic networks, because it involves many fewer reactions than carbon metabolism. Specifically, we study properties of the space of all possible metabolic genotypes, and analyze properties of random metabolic genotypes that are viable on different numbers of sulfur sources. We show that metabolic genotypes with the same phenotype form large connected genotype networks that extend far through metabolic genotype space. How far they reach through this space is a linear function of the number of super-essential reactions in such networks, the number of reactions that occur in all networks with the same phenotype. We show that different neighborhoods of any genotype network harbor very different novel phenotypes, metabolic innovations that can sustain life on novel sulfur sources. We also analyze the ability of evolving populations of metabolic networks to explore novel metabolic phenotypes. This ability is facilitated by the existence of genotype networks, because different neighborhoods of these networks contain very different novel phenotypes. In contrast to macromolecules, where phenotypic robustness may facilitate phenotypic innovation, we show that here the ability to access novel phenotypes does not monotonically increase with robustness.

32 citations


Posted Content
TL;DR: The results validate and explain the ubiquitous appearance of the quadratic scaling for a broad spectrum of topologies of underlying universal metabolic networks and demonstrate why, in spite of “small-world” topology, real-life metabolic networks are characterized by a broad distribution of pathway lengths and sizes in regulatory networks.
Abstract: In prokaryotic genomes the number of transcriptional regulators is known to quadratically scale with the total number of protein-coding genes. Toolbox model was recently proposed to explain this scaling for metabolic enzymes and their regulators. According to its rules the metabolic network of an organism evolves by horizontal transfer of pathways from other species. These pathways are part of a larger "universal" network formed by the union of all species-specific networks. It remained to be understood, however, how the topological properties of this universal network influence the scaling law of functional content of genomes. In this study we answer this question by first analyzing the scaling properties of the toolbox model on arbitrary tree-like universal networks. We mathematically prove that the critical branching topology, in which the average number of upstream neighbors of a node is equal to one, is both necessary and sufficient for the quadratic scaling. Conversely, the toolbox model on trees with exponentially expanding, supercritical topology is characterized by the linear scaling with logarithmic corrections. We further generalize our model to include reactions with multiple substrates/products as well as branched or cyclic metabolic pathways. Unlike the original model the new version employs evolutionary optimized pathways with the smallest number of reactions necessary to achieve their metabolic tasks. Numerical simulations of this most realistic model on the universal network from the KEGG database again produced approximately quadratic scaling. Our results demonstrate why, in spite of their "small-world" topology, real-life metabolic networks are characterized by a broad distribution of pathway lengths and sizes of metabolic regulons in regulatory networks.

22 citations


Journal ArticleDOI
TL;DR: It is shown that examples of catalytic networks include synthetic DNA molecular circuits that have been shown to perform signal amplification and molecular logic and the main theorem is that all weakly-reversible networks with critical siphons are catalytic.
Abstract: We define catalytic networks as chemical reaction networks with an essentially catalytic reaction pathway: one which is on in the presence of certain catalysts and off in their absence. We show that examples of catalytic networks include synthetic DNA molecular circuits that have been shown to perform signal amplification and molecular logic. Recall that a critical siphon is a subset of the species in a chemical reaction network whose absence is forward invariant and stoichiometrically compatible with a positive point. Our main theorem is that all weakly-reversible networks with critical siphons are catalytic. Consequently, we obtain new proofs for the persistence of atomic event-systems of Adleman et al., and normal networks of Gnacadja. We define autocatalytic networks, and conjecture that a weakly-reversible reaction network has critical siphons if and only if it is autocatalytic.

19 citations


Posted Content
TL;DR: In this paper, the authors derived conditions based on linear inequalities that allow the analytic computation of states and parameters where the Jacobian derived from a mass action network has a defective zero eigenvalue so that -- under certain genericity conditions -- a saddle-node bifurcation occurs.
Abstract: Many biochemical processes can successfully be described by dynamical systems allowing some form of switching when, depending on their initial conditions, solutions of the dynamical system end up in different regions of state space (associated with different biochemical functions). Switching is often realized by a bistable system (i.e. a dynamical system allowing two stable steady state solutions) and, in the majority of cases, bistability is established numerically. In our point of view this approach is too restrictive, as, one the one hand, due to predominant parameter uncertainty numerical methods are generally difficult to apply to realistic models originating in Systems Biology. And on the other hand switching already arises with the occurrence of a saddle type steady state (characterized by a Jacobian where exactly one Eigenvalue is positive and the remaining eigenvalues have negative real part). Consequently we derive conditions based on linear inequalities that allow the analytic computation of states and parameters where the Jacobian derived from a mass action network has a defective zero eigenvalue so that -- under certain genericity conditions -- a saddle-node bifurcation occurs. Our conditions are applicable to general mass action networks involving at least one conservation relation, however, they are only sufficient (as infeasibility of linear inequalities does not exclude defective zero eigenvalues).

14 citations


Journal ArticleDOI
TL;DR: A characterization of the productive capabilities of the metabolic network of the bacterium E.coli in a specified growth medium in terms of the producible biochemical species is provided.
Abstract: Flux analysis is a class of constraint-based approaches to the study of biochemical reaction networks: they are based on determining the reaction flux configurations compatible with given stoichiometric and thermodynamic constraints. One of its main areas of application is the study of cellular metabolic networks. We briefly and selectively review the main approaches to this problem and then, building on recent work, we provide a characterization of the productive capabilities of the metabolic network of the bacterium E.coli in a specified growth medium in terms of the producible biochemical species. While a robust and physiologically meaningful production profile clearly emerges (including biomass components, biomass products, waste etc.), the underlying constraints still allow for significant fluctuations even in key metabolites like ATP and, as a consequence, apparently lay the ground for very different growth scenarios.

12 citations


Book ChapterDOI
TL;DR: This chapter focuses its attention on a particular class of methods for reverse-engineering, namely those that rely algorithmically upon the so-called "hitting-set" problem, which is a classical combinatorial and computer science problem.
Abstract: The understanding of molecular cell biology requires insight into the structure and dynamics of networks that are made up of thousands of interacting molecules of DNA, RNA, proteins, metabolites, and other components. One of the central goals of systems biology is the unraveling of the as yet poorly characterized complex web of interactions among these components. This work is made harder by the fact that new species and interactions are continuously discovered in experimental work, necessitating the development of adaptive and fast algorithms for network construction and updating. Thus, the "reverse-engineering" of networks from data has emerged as one of the central concern of systems biology research. A variety of reverse-engineering methods have been developed, based on tools from statistics, machine learning, and other mathematical domains. In order to effectively use these methods, it is essential to develop an understanding of the fundamental characteristics of these algorithms. With that in mind, this chapter is dedicated to the reverse-engineering of biological systems. Specifically, we focus our attention on a particular class of methods for reverse-engineering, namely those that rely algorithmically upon the so-called "hitting-set" problem, which is a classical combinatorial and computer science problem, Each of these methods utilizes a different algorithm in order to obtain an exact or an approximate solution of the hitting set problem. We will explore the ultimate impact that the alternative algorithms have on the inference of published in silico biological networks.

Posted Content
TL;DR: Monte Carlo simulations of apoptotic signaling provides unexpected insights into the mechanisms of fractional cell killing induced by apoptosis-inducing agents, showing that not only variation in protein levels, but also inherent stochastic variability in signaling reactions, can lead to survival of a fraction of treated cancer cells.
Abstract: Cancer cells are widely known to be protected from apoptosis, which is a major hurdle to successful anti-cancer therapy. Over-expression of several anti-apoptotic proteins, or mutations in pro-apoptotic factors, has been recognized to confer such resistance. Development of new experimental strategies, such as in silico modeling of biological pathways, can increase our understanding of how abnormal regulation of apoptotic pathway in cancer cells can lead to tumour chemoresistance. Monte Carlo simulations are in particular well suited to study inherent variability, such as spatial heterogeneity and cell-to-cell variations in signaling reactions. Using this approach, often in combination with experimental validation of the computational model, we observed that large cell-to-cell variability could explain the kinetics of apoptosis, which depends on the type of pathway and the strength of stress stimuli. Most importantly, Monte Carlo simulations of apoptotic signaling provides unexpected insights into the mechanisms of fractional cell killing induced by apoptosis-inducing agents, showing that not only variation in protein levels, but also inherent stochastic variability in signaling reactions, can lead to survival of a fraction of treated cancer cells.

Journal ArticleDOI
TL;DR: In this paper, the authors used probabilistic computational modeling of the mitochondrial pathway of apoptosis, verified by single-cell experimental observations, to develop a model of Bcl-2 inhibition of cancer cells.
Abstract: BH3 mimetics have been proposed as new anticancer therapeutics. They target anti-apoptotic Bcl-2 proteins, up-regulation of which has been implicated in the resistance of many cancer cells, particularly leukemia and lymphoma cells, to apoptosis. Using probabilistic computational modeling of the mitochondrial pathway of apoptosis, verified by single-cell experimental observations, we develop a model of Bcl-2 inhibition of apoptosis. Our results clarify how Bcl-2 imparts its anti-apoptotic role by increasing the time-to-death and cell-to-cell variability. We also show that although the commitment to death is highly impacted by differences in protein levels at the time of stimulation, inherent stochastic fluctuations in apoptotic signaling are sufficient to induce cell-to-cell variability and to allow single cells to escape death. This study suggests that intrinsic cell-to-cell stochastic variability in apoptotic signaling is sufficient to cause fractional killing of cancer cells after exposure to BH3 mimetics. This is an unanticipated facet of cancer chemoresistance.

Journal ArticleDOI
TL;DR: It is argued that the primary role of positive thymic selection and the resulting T cell population is the maintenance of a homeostatic equilibrium with self MHC-self peptide complexes.
Abstract: Our main tenet argues that the primary role of positive thymic selection and the resulting T cell population is the maintenance of a homeostatic equilibrium with self MHC-self peptide complexes. The homeostatic T cell repertoire can recognize infections non-specifically and this is an indirect (negative) recognition: the whole homeostatic T cell population together "holds a mirror" to the whole self, and any MHC-peptide complex that is "not reflected in the mirror" can be perceived by surrounding homeostatic T cells as a signal of the presence of a foreign entity. On the other hand, infection-specific T cell clones arise in a different pathway in the periphery, do not enter the thymus, and form a functionally different population. Here we summarize the basic assumptions and consequences of a logic-based new model, which differs from conventional models in many respects.

Posted Content
TL;DR: A network-based scoring system of reaction scoring that exploits the complex hierarchical structure and the statistical regularities of the metabolic network as a bipartite graph is proposed and, after validation, individual link information is integrated to assess the reliability of each reaction in probabilistic terms.
Abstract: Reliability on complex biological networks reconstructions remains a concern. Although observations are getting more and more precise, the data collection process is yet error prone and the proofs display uneven certitude. In the case of metabolic networks, the currently employed confidence scoring system rates reactions according to a discretized small set of labels denoting different levels of experimental evidence or model-based likelihood. Here, we propose a computational network-based system of reaction scoring that exploits the complex hierarchical structure and the statistical regularities of the metabolic network as a bipartite graph. We use the example of Escherichia coli metabolism to illustrate our methodology. Our model is adjusted to the observations in order to derive connection probabilities between individual metabolite-reaction pairs and, after validation, we integrate individual link information to assess the reliability of each reaction in probabilistic terms. This network-based scoring system breaks the degeneracy of currently employed scores, enables further confirmation of modeling results, uncovers very specific reactions that could be functionally or evolutionary important, and identifies prominent experimental targets for further verification. We foresee a wide range of potential applications of our approach given the natural network bipartivity of many biological interactions.

Posted Content
TL;DR: These are notes for a set of 7 two-hour lectures given at the 2010 Summer School on Quantitative Evolutionary and Comparative Genomics at OIST, Okinawa, Japan, with the emphasis on understanding how biological systems process information.
Abstract: These are notes for a set of 7 two-hour lectures given at the 2010 Summer School on Quantitative Evolutionary and Comparative Genomics at OIST, Okinawa, Japan. The emphasis is on understanding how biological systems process information. We take a physicist's approach of looking for simple phenomenological descriptions that can address the questions of biological function without necessarily modeling all (mostly unknown) microscopic details; the example that is developed throughout the notes is transcriptional regulation in genetic regulatory networks. We present tools from information theory and statistical physics that can be used to analyze noisy nonlinear biological networks, and build generative and predictive models of regulatory processes.

Posted Content
TL;DR: RNAnet as discussed by the authors provides a bridge between two widely used Human gene databases, Ensembl describes DNA sequences and transcripts but not experimental gene expression. But without RNAnet comparison across experiments in GEO is very labour intensive requiring man months of effort to down load and clean data.
Abstract: RNAnet provides a bridge between two widely used Human gene databases. Ensembl describes DNA sequences and transcripts but not experimental gene expression. Whilst NCBI's GEO contains actual expression levels from Human samples. RNAnet provides immediate access to thousands of Affymetrix HG-U133 2plus GeneChip measurements Homo sapiens genes in most medically interesting tissues. Without RNAnet comparison across experiments in GEO is very labour intensive requiring man months of effort to down load and clean data. With RNAnet anyone can access cleaned quantile normalised data in seconds. It can be used to data mine patterns of co-expression. The network of strongly correlated genes is huge but sparse. Thousands of genes interact strongly with thousands of others. Conversely there are tens of thousands of genes which interact strongly with less than 100 others. I.e. RNAnet gives new views for RNA Systems Biology.

Journal ArticleDOI
TL;DR: Using cell biology and biochemical approaches, it is demonstrated that neuroglobin inhibits the intrinsic pathway of apoptosis in vitro and intervenes in activation of pro-caspase 9 by interaction with cytochrome c and this model provides an explanation the action of Neuroglobin in the protection of nerve cells from unwanted apoptosis.
Abstract: In the past few years, overwhelming evidence has accrued that a high level of expression of the protein neuroglobin protects neurons in vitro, in animal models, and in humans, against cell death associated with hypoxic and amyloid insult. However, until now, the exact mechanism of neuroglobin's protective action has not been determined. Using cell biology and biochemical approaches we demonstrate that neuroglobin inhibits the intrinsic pathway of apoptosis in vitro and intervenes in activation of pro-caspase 9 by interaction with cytochrome c. Using systems level information of the apoptotic signalling reactions we have developed a quantitative model of neuroglobin inhibition of apoptosis, which simulates neuroglobin blocking of apoptosome formation at a single cell level. Furthermore, this model allows us to explore the effect of neuroglobin in conditions not easily accessible to experimental study. We found that the protection of neurons by neuroglobin is very concentration sensitive. The impact of neuroglobin may arise from both its binding to cytochrome c and its subsequent redox reaction, although the binding alone is sufficient to block pro-caspase 9 activation. These data provides an explanation the action of neuroglobin in the protection of nerve cells from unwanted apoptosis.

Posted Content
TL;DR: Analysis of a comprehensive data set of protein-protein and transcriptional regulatory interaction networks in yeast, an Escherichia coli metabolic network, and gene activity profiles for different metabolic states shows that in all cases the networks have a heavy-tailed distribution.
Abstract: Various molecular interaction networks have been claimed to follow power-law decay for their global connectivity distribution. It has been proposed that there may be underlying generative models that explain this heavy-tailed behavior by self-reinforcement processes such as classical or hierarchical scale-free network models. Here we analyze a comprehensive data set of protein-protein and transcriptional regulatory interaction networks in yeast, an E. coli metabolic network, and gene activity profiles for different metabolic states in both organisms. We show that in all cases the networks have a heavy-tailed distribution, but most of them present significant differences from a power-law model according to a stringent statistical test. Those few data sets that have a statistically significant fit with a power-law model follow other distributions equally well. Thus, while our analysis supports that both global connectivity interaction networks and activity distributions are heavy-tailed, they are not generally described by any specific distribution model, leaving space for further inferences on generative models.

Journal ArticleDOI
TL;DR: In this paper, the extrinsic noise is regarded as fluctuations in the values of kinetic parameters and such fluctuations are modeled by randomly sampling the kinetic rate constants from a uniform distribution.
Abstract: Cellular responses in the single cells are known to be highly heterogeneous and individualistic due to the strong influence by extrinsic and intrinsic noise. Here, we are concerned about how to model the extrinsic noise-induced heterogeneous response in the single cells under the constraints of experimentally obtained population-averaged response, but without much detailed kinetic information. We propose a novel statistical ensemble scheme where extrinsic noise is regarded as fluctuations in the values of kinetic parameters and such fluctuations are modeled by randomly sampling the kinetic rate constants from a uniform distribution. We consider a large number of signaling system replicates, each of which has the same network topology, but a uniquely different set of kinetic rate constants. A protein dynamic response from each replicate should represent the dynamics in a single cell and the statistical ensemble average should be regarded as a population-level response averaged over a population of the cells. We devise an optimization algorithm to find the correct uniform distribution of the network parameters, which produces the correct statistical distribution of the response whose ensemble average and distribution agree well with the population-level experimental data and the experimentally observed heterogeneity. We apply this statistical ensemble analysis to a NF-{\kappa}B signaling system and (1) predict the distributions of the heterogeneous NF-{\kappa}B (either oscillatory or non-oscillatory) dynamic patterns and of the dynamic features (e.g., period), (2) predict that both the distribution and the statistical ensemble average of the NF-{\kappa}B dynamic response depends sensitively on the dosage of stimulant, and lastly (3) demonstrate the sigmoidally shaped dose-response from the statistical ensemble average and the individual replicates.

Posted Content
TL;DR: An overview of the spectral distances is shown, highlighting their behavior in some basic cases of static and dynamic synthetic and real networks.
Abstract: Many functions have been recently defined to assess the similarity among networks as tools for quantitative comparison. They stem from very different frameworks - and they are tuned for dealing with different situations. Here we show an overview of the spectral distances, highlighting their behavior in some basic cases of static and dynamic synthetic and real networks.

Posted Content
TL;DR: Dynamical processes occurring at the two different length scales of within the cell and between cells, viz., the intra-cellular signaling network and the nervous system are described, showing that focusing on the systems-level aspects of these problems allows one to observe surprising and illuminating common themes amongst them.
Abstract: With the completion of human genome mapping, the focus of scientists seeking to explain the biological complexity of living systems is shifting from analyzing the individual components (such as a particular gene or biochemical reaction) to understanding the set of interactions amongst the large number of components that results in the different functions of the organism. To this end, the area of systems biology attempts to achieve a "systems-level" description of biology by focusing on the network of interactions instead of the characteristics of its isolated parts. In this article, we briefly describe some of the emerging themes of research in "network" biology, looking at dynamical processes occurring at the two different length scales of within the cell and between cells, viz., the intra-cellular signaling network and the nervous system. We show that focusing on the systems-level aspects of these problems allows one to observe surprising and illuminating common themes amongst them.

Journal ArticleDOI
TL;DR: In this paper, the authors focus on the Cad module, a subsystem of E. coli's response to acidic stress, which is conditionally activated at low pH only when lysine is available.
Abstract: The analysis of stress response systems in microorganisms can reveal molecular strategies for regulatory control and adaptation. Here, we focus on the Cad module, a subsystem of E. coli's response to acidic stress, which is conditionally activated at low pH only when lysine is available. When expressed, the Cad system counteracts the elevated H+ concentration by converting lysine to cadaverine under the consumption of H+, and exporting cadaverine in exchange for external lysine. Surprisingly, the cad operon displays a transient response, even when the conditions for its induction persist. To quantitatively characterize the regulation of the Cad module, we have experimentally recorded and theoretically modeled the dynamics of important system variables. We establish a quantitative model that adequately describes and predicts the transient expression behavior for various initial conditions. Our quantitative analysis of the Cad system supports a negative feedback by external cadaverine as the origin of the transient response. Furthermore, the analysis puts causal constraints on the precise mechanism of signal transduction via the regulatory protein CadC.

Journal ArticleDOI
TL;DR: How rescue interactions can lead to the rational design of antagonistic drug combinations that select against resistance and how they can illuminate medical research on cancer, antibiotics, and metabolic diseases are discussed.
Abstract: Recent research shows that a faulty or sub-optimally operating metabolic network can often be rescued by the targeted removal of enzyme-coding genes--the exact opposite of what traditional gene therapy would suggest. Predictions go as far as to assert that certain gene knockouts can restore the growth of otherwise nonviable gene-deficient cells. Many questions follow from this discovery: What are the underlying mechanisms? How generalizable is this effect? What are the potential applications? Here, I will approach these questions from the perspective of compensatory perturbations on networks. Relations will be drawn between such synthetic rescues and naturally occurring cascades of reaction inactivation, as well as their analogues in physical and other biological networks. I will specially discuss how rescue interactions can lead to the rational design of antagonistic drug combinations that select against resistance and how they can illuminate medical research on cancer, antibiotics, and metabolic diseases.

Posted Content
TL;DR: Stability analysis and simulation of SLIMI mathematical models show that this lateral inhibition circuit is capable of pattern formation across a broad range of parameter values, and provides a simple and plausible explanation for lateral inhibition pattern formation during development.
Abstract: Lateral inhibition patterns mediated by the Notch-Delta signaling system occur in diverse developmental contexts These systems are based on an intercellular feedback loop in which Notch activation leads to down-regulation of Delta However, even in relatively well-characterized systems, the pathway leading from Notch activation to Delta repression often remains elusive Recent work has shown that cis-interactions between Notch and Delta lead to mutual inactivation of both proteins Here we show that this type of cis-interaction enables a simpler and more direct mechanism for lateral inhibition feedback than those proposed previously In this mechanism, Notch signaling directly up-regulates Notch expression, thereby inactivating Delta through the mutual inactivation of Notch and Delta proteins This mechanism, which we term Simplest Lateral Inhibition by Mutual Inactivation (SLIMI), can implement patterning without requiring any additional genes or regulatory interactions Moreover, the key interaction of Notch expression in response to Notch signaling has been observed in some systems Stability analysis and simulation of SLIMI mathematical models show that this lateral inhibition circuit is capable of pattern formation across a broad range of parameter values These results provide a simple and plausible explanation for lateral inhibition pattern formation during development

Posted Content
TL;DR: It is shown that networks with an inhomogeneous degree distribution are more robust to crosstalk than corresponding homogeneous networks and an algorithm to find the optimal interconnection structure among their vertices is presented.
Abstract: Crosstalk is defined as the set of unwanted interactions among the different entities of a network. Crosstalk is present in various degrees in every system where information is transmitted through a means that is accessible by all the individual units of the network. Using concepts from graph theory, we introduce a quantifiable measure for sensitivity to crosstalk, and analytically derive the structure of the networks in which it is minimized. It is shown that networks with an inhomogeneous degree distribution are more robust to crosstalk than corresponding homogeneous networks. We provide a method to construct the graph with the minimum possible sensitivity to crosstalk, given its order and size. Finally, for networks with a fixed degree sequence, we present an algorithm to find the optimal interconnection structure among their vertices.

Posted Content
TL;DR: It is shown that the graph structure of the realization containing the maximal number of reactions is unique if the set of possible complexes is fixed and a mixed integer programming based numerical procedure is given for computing a realizing containing the minimal/maximal number of complexes.
Abstract: This paper presents new results about the optimization based generation of chemical reaction networks (CRNs) of higher deficiency. Firstly, it is shown that the graph structure of the realization containing the maximal number of reactions is unique if the set of possible complexes is fixed. Secondly, a mixed integer programming based numerical procedure is given for computing a realization containing the minimal/maximal number of complexes. Moreover, the linear inequalities corresponding to full reversibility of the CRN realization are also described. The theoretical results are illustrated on meaningful examples.

Posted Content
TL;DR: Higher order Boolean networks are introduced, which feature an explicit distinction between the different cell components and the types of interactions between them, and it is shown that the stability of the cell state dynamics can be determined solving the eigenvalue problem of a matrix representing the regulatory interactions and their strengths.
Abstract: The regulation of the cell state is a complex process involving several components. These complex dynamics can be modeled using Boolean networks, allowing us to explain the existence of different cell states and the transition between them. Boolean models have been introduced both as specific examples and as ensemble or distribution network models. However, current ensemble Boolean network models do not make a systematic distinction between different cell components such as epigenetic factors, gene and transcription factors. Consequently, we still do not understand their relative contributions in controlling the cell fate. In this work we introduce and study higher order Boolean networks, which feature an explicit distinction between the different cell components and the types of interactions between them. We show that the stability of the cell state dynamics can be determined solving the eigenvalue problem of a matrix representing the regulatory interactions and their strengths. The qualitative analysis of this problem indicates that, in addition to the classification into stable and chaotic regimes, the cell state can be simple or complex depending on whether it can be deduced from the independent study of its components or not. Finally, we illustrate how the model can be expanded considering higher levels and higher order dynamics.

Journal ArticleDOI
TL;DR: In this paper, the authors reveal a systemic virus-entry network initiated by human cytomegalovirus (HCMV), a widespread opportunistic pathogen, which contains all known interactions and functional modules (i.e. groups of proteins) coordinately responding to HCMV entry.
Abstract: Virus entry is a multistep process that triggers a variety of cellular pathways interconnecting into a complex network, yet the molecular complexity of this network remains largely unsolved. Here, by employing systems biology approach, we reveal a systemic virus-entry network initiated by human cytomegalovirus (HCMV), a widespread opportunistic pathogen. This network contains all known interactions and functional modules (i.e. groups of proteins) coordinately responding to HCMV entry. The number of both genes and functional modules activated in this network dramatically declines shortly, within 25 min post-infection. While modules annotated as receptor system, ion transport, and immune response are continuously activated during the entire process of HCMV entry, those for cell adhesion and skeletal movement are specifically activated during viral early attachment, and those for immune response during virus entry. HCMV entry requires a complex receptor network involving different cellular components, comprising not only cell surface receptors, but also pathway components in signal transduction, skeletal development, immune response, endocytosis, ion transport, macromolecule metabolism and chromatin remodeling. Interestingly, genes that function in chromatin remodeling are the most abundant in this receptor system, suggesting that global modulation of transcriptions is one of the most important events in HCMV entry. Results of in silico knock out further reveal that this entire receptor network is primarily controlled by multiple elements, such as EGFR (Epidermal Growth Factor) and SLC10A1 (sodium/bile acid cotransporter family, member 1). Thus, our results demonstrate that a complex systemic network, in which components coordinating efficiently in time and space contributes to virus entry.

Posted Content
TL;DR: A report of the meeting "Challenges in experimental data integration within genome-scale metabolic models", Institut Henri Poincaré, Paris, October 10-11 2009, organized by the CNRS-MPG joint program in Systems Biology.
Abstract: A report of the meeting "Challenges in experimental data integration within genome-scale metabolic models", Institut Henri Poincar\'e, Paris, October 10-11 2009, organized by the CNRS-MPG joint program in Systems Biology.