scispace - formally typeset
Search or ask a question

Showing papers in "arXiv: Molecular Networks in 2014"


Journal ArticleDOI
TL;DR: In this article, a cancer hallmark network framework for modelling genome-sequencing data to predict cancer clonal evolution and associated clinical phenotypes is discussed. And the authors discuss strategies of using this framework in conjunction with genome sequencing data in an attempt to predict personalized drug targets, drug resistance, and metastasis for a cancer patient, as well as cancer risks for a healthy individual.
Abstract: We discuss a cancer hallmark network framework for modelling genome-sequencing data to predict cancer clonal evolution and associated clinical phenotypes. Strategies of using this framework in conjunction with genome sequencing data in an attempt to predict personalized drug targets, drug resistance, and metastasis for a cancer patient, as well as cancer risks for a healthy individual are discussed. Accurate prediction of cancer clonal evolution and clinical phenotypes will have substantial impact on timely diagnosis, personalized management and prevention of cancer.

152 citations


Journal ArticleDOI
TL;DR: Due to its novel features, ComPPI is useful for the analysis of experimental results in biochemistry and molecular biology, as well as for proteome-wide studies in bioinformatics and network science helping cellular biology, medicine and drug design.
Abstract: Here we present ComPPI, a cellular compartment specific database of proteins and their interactions enabling an extensive, compartmentalized protein-protein interaction network analysis (this http URL). ComPPI enables the user to filter biologically unlikely interactions, where the two interacting proteins have no common subcellular localizations and to predict novel properties, such as compartment-specific biological functions. ComPPI is an integrated database covering four species (S. cerevisiae, C. elegans, D. melanogaster and H. sapiens). The compilation of nine protein-protein interaction and eight subcellular localization data sets had four curation steps including a manually built, comprehensive hierarchical structure of more than 1600 subcellular localizations. ComPPI provides confidence scores for protein subcellular localizations and protein-protein interactions. ComPPI has user-friendly search options for individual proteins giving their subcellular localization, their interactions and the likelihood of their interactions considering the subcellular localization of their interacting partners. Download options of search results, whole proteomes, organelle-specific interactomes and subcellular localization data are available on its website. Due to its novel features, ComPPI is useful for the analysis of experimental results in biochemistry and molecular biology, as well as for proteome-wide studies in bioinformatics and network science helping cellular biology, medicine and drug design.

87 citations


Journal ArticleDOI
TL;DR: A tumor often consists of multiple cell subpopulations (clones), and current chemo-treatments often target one clone of a tumor, other clones overtake it and the tumor reoccurs.
Abstract: A tumor often consists of multiple cell subpopulations (clones). Current chemo-treatments often target one clone of a tumor. Although the drug kills that clone, other clones overtake it and the tumor reoccurs. Genome sequencing and computational analysis allows to computational dissection of clones from tumors, while singe-cell genome sequencing including RNA-Seq allows to profiling of these clones. This opens a new window for treating a tumor as a system in which clones are evolving. Future cancer systems biology studies should consider a tumor as an evolving system with multiple clones. Therefore, topics discussed in Part 2 of this review include evolutionary dynamics of clonal networks, early-warning signals for formation of fast-growing clones, dissecting tumor heterogeneity, and modeling of clone-clone-stroma interactions for drug resistance. The ultimate goal of the future systems biology analysis is to obtain a whole-system understanding of a tumor and therefore provides a more efficient and personalized management strategies for cancer patients.

55 citations


Journal ArticleDOI
TL;DR: It is shown that for bistable and oscillatory chemical systems with deterministic initial conditions, the solution of the MA equations can be interpreted as a valid approximation to the true moments of the chemical master equation, only when the steady-state mean molecule numbers obtained from thechemical master equation fall within a certain finite range.
Abstract: Approximations based on moment-closure (MA) are commonly used to obtain estimates of the mean molecule numbers and of the variance of fluctuations in the number of molecules of chemical systems. The advantage of this approach is that it can be far less computationally expensive than exact stochastic simulations of the chemical master equation. Here we numerically study the conditions under which the MA equations yield results reflecting the true stochastic dynamics of the system. We show that for bistable and oscillatory chemical systems with deterministic initial conditions, the solution of the MA equations can be interpreted as a valid approximation to the true moments of the CME, only when the steady-state mean molecule numbers obtained from the chemical master equation fall within a certain finite range. The same validity criterion for monostable systems implies that the steady-state mean molecule numbers obtained from the chemical master equation must be above a certain threshold. For mean molecule numbers outside of this range of validity, the MA equations lead to either qualitatively wrong oscillatory dynamics or to unphysical predictions such as negative variances in the molecule numbers or multiple steady-state moments of the stationary distribution as the initial conditions are varied. Our results clarify the range of validity of the MA approach and show that pitfalls in the interpretation of the results can only be overcome through the systematic comparison of the solutions of the MA equations of a certain order with those of higher orders.

53 citations


Posted Content
TL;DR: It is shown that Shannon's information entropy, compressibility and algorithmic complexity quantify different local and global aspects of synthetic and biological data, and it is proved that the Kolmogorov complexity of a labeled graph is a good approximation of its unlabeled Kolmogsorv complexity and thus a robust definition of graph complexity.
Abstract: We survey and introduce concepts and tools located at the intersection of information theory and network biology. We show that Shannon's information entropy, compressibility and algorithmic complexity quantify different local and global aspects of synthetic and biological data. We show examples such as the emergence of giant components in Erdos-Renyi random graphs, and the recovery of topological properties from numerical kinetic properties simulating gene expression data. We provide exact theoretical calculations, numerical approximations and error estimations of entropy, algorithmic probability and Kolmogorov complexity for different types of graphs, characterizing their variant and invariant properties. We introduce formal definitions of complexity for both labeled and unlabeled graphs and prove that the Kolmogorov complexity of a labeled graph is a good approximation of its unlabeled Kolmogorov complexity and thus a robust definition of graph complexity.

53 citations


Journal ArticleDOI
TL;DR: In this article, a mathematical criterion based on the topological features of the transcriptional regulatory networks (TRNs) was proposed to classify the network elements into one of two possible classes: hierarchical or modular genes.
Abstract: Background: Previous studies have used different methods in an effort to extract the modular organization of transcriptional regulatory networks (TRNs). However, these approaches are not natural, as they try to cluster highly connected genes into a module or locate known pleiotropic transcription factors in lower hierarchical layers. Here, we unravel the TRN of Escherichia coli by separating it into its key elements, thus revealing its natural organization. We also present a mathematical criterion, based on the topological features of the TRN, to classify the network elements into one of two possible classes: hierarchical or modular genes. Results: We found that modular genes are clustered into physiologically correlated groups validated by a statistical analysis of the enrichment of the functional classes. Hierarchical genes encode transcription factors responsible for coordinating module responses based on general interest signals. Hierarchical elements correlate highly with the previously studied global regulators, suggesting that this could be the first mathematical method to identify global regulators. We identified a new element in TRNs never described before: intermodular genes. These are structural genes that integrate, at the promoter level, signals coming from different modules, and therefore from different physiological responses. Using the concept of pleiotropy, we have reconstructed the hierarchy of the network and discuss the role of feedforward motifs in shaping the hierarchical backbone of the TRN. Conclusions: This study sheds new light on the design principles underpinning the organization of TRNs, showing a novel nonpyramidal architecture composed of independent modules globally governed by hierarchical transcription factors, whose responses are integrated by intermodular genes.

40 citations


Posted Content
TL;DR: FUSE as mentioned in this paper is a multiple network aligner that utilizes all functional and topological information in all PPI networks by fusing from all aligned networks both the protein wiring patterns and their sequence similarities.
Abstract: Discovering patterns in networks of protein-protein interactions (PPIs) is a central problem in systems biology. Alignments between these networks aid functional understanding as they uncover important information, such as evolutionary conserved pathways, protein complexes and functional orthologs. The objective of a multiple network alignment is to create clusters of nodes that are evolutionarily conserved and functionally consistent across all networks. Unfortunately, the alignment methods proposed thus far do not fully meet this objective, as they are guided by pairwise scores that do not utilize the entire functional and topological information across all networks. To overcome this weakness, we propose FUSE, a multiple network aligner that utilizes all functional and topological information in all PPI networks. It works in two steps. First, it computes novel similarity scores of proteins across the PPI networks by fusing from all aligned networks both the protein wiring patterns and their sequence similarities. It does this by using Non-negative Matrix Tri-Factorization (NMTF). When we apply NMTF on the five largest and most complete PPI networks from BioGRID, we show that NMTF finds a larger number of protein pairs across the PPI networks that are functionally conserved than can be found by using protein sequence similarities alone. This demonstrates complementarity of protein sequence and their wiring patterns in the PPI networks. In the second step, FUSE uses a novel maximum weight k-partite matching approximation algorithm to find an alignment between multiple networks. We compare FUSE with the state of the art multiple network aligners and show that it produces the largest number of functionally consistent clusters that cover all aligned PPI networks. Also, FUSE is more computationally efficient than other multiple network aligners.

36 citations


Journal ArticleDOI
TL;DR: A novel approach to drug target deconvolution compares the abundance profiles of proteins expressed in a panel of cells treated with different drugs, and identifies proteins with cell-type independent and drug-specific regulation that is exceptionally strong in relation to the other proteins.
Abstract: Phenomenological screening of small molecule libraries for anticancer activity yields potentially interesting candidate molecules, with a bottleneck in the determination of drug targets and the mechanism of anticancer action. A novel approach to drug target deconvolution compares the abundance profiles of proteins expressed in a panel of cells treated with different drugs, and identifies proteins with cell-type independent and drug-specific regulation that is exceptionally strong in relation to the other proteins. Mapping top candidates on known protein networks reveals the mechanism of drug action, while abundant proteins provide a signature of cellular death/survival pathways. The above approach can significantly shorten drug target identification, and thus facilitate the emergence of novel anticancer treatments.

31 citations


Journal ArticleDOI
TL;DR: A quantitative bound on the error between the pathwise realizations of these two processes is proved, providing a foundation for using delay stochastic differential equations to approximate the dynamics of birth-death processes with delay.
Abstract: Delay is an important and ubiquitous aspect of many biochemical processes. For example, delay plays a central role in the dynamics of genetic regulatory networks as it stems from the sequential assembly of first mRNA and then protein. Genetic regulatory networks are therefore frequently modeled as stochastic birth-death processes with delay. Here we examine the relationship between delay birth-death processes and their appropriate approximating delay chemical Langevin equations. We prove that the distance between these two descriptions, as measured by expectations of functionals of the processes, converges to zero with increasing system size. Further, we prove that the delay birth-death process converges to the thermodynamic limit as system size tends to infinity. Our results hold for both fixed delay and distributed delay. Simulations demonstrate that the delay chemical Langevin approximation is accurate even at moderate system sizes. It captures dynamical features such as the spatial and temporal distributions of transition pathways in metastable systems, oscillatory behavior in negative feedback circuits, and cross-correlations between nodes in a network. Overall, these results provide a foundation for using delay stochastic differential equations to approximate the dynamics of birth-death processes with delay.

29 citations


Journal ArticleDOI
TL;DR: FALCON as mentioned in this paper employs metabolic network reconstructions along with expression data to estimate fluxes, which can be used to increase predictive capability resulting in improved understanding of metabolism, but it does not provide an objective function for the estimation of fluxes.
Abstract: A major theme in constraint-based modeling is unifying experimental data, such as biochemical information about the reactions that can occur in a system or the composition and localization of enzyme complexes, with highthroughput data including expression data, metabolomics, or DNA sequencing. The desired result is to increase predictive capability resulting in improved understanding of metabolism. The approach typically employed when only gene (or protein) intensities are available is the creation of tissue-specific models, which reduces the available reactions in an organism model, and does not provide an objective function for the estimation of fluxes, which is an important limitation in many modeling applications. We develop a method, flux assignment with LAD (least absolute deviation) convex objectives and normalization (FALCON), that employs metabolic network reconstructions along with expression data to estimate fluxes. In order to use such a method, accurate measures of enzyme complex abundance are needed, so we first present a new algorithm that addresses quantification of complex abundance. Our extensions to prior techniques include the capability to work with large models and significantly improved run-time performance even for smaller models, an improved analysis of enzyme complex formation logic, the ability to handle very large enzyme complex rules that may incorporate multiple isoforms, and depending on the model constraints, either maintained or significantly improved correlation with experimentally measured fluxes. FALCON has been implemented in MATLAB and ATS, and can be downloaded from: this https URL ATS is not required to compile the software, as intermediate C source code is available, and binaries are provided for Linux x86-64 systems. FALCON requires use of the COBRA Toolbox, also implemented in MATLAB.

26 citations


Journal ArticleDOI
TL;DR: The analysis of the breast cancer network and its normal counterpart at the proteomic level provides a benchmark for designing drugs which can target a subgraph instead of individual proteins.
Abstract: Breast cancer has been reported to account for the maximum cases among all female cancers till date. In order to gain a deeper insight into the complexities of the disease, we analyze the breast cancer network and its normal counterpart at the proteomic level. While the short range correlations in the eigenvalues exhibiting universality provide an evidence towards the importance of random connections in the underlying networks, the long range correlations along with the localization properties reveal insightful structural patterns involving functionally important proteins. The analysis provides a benchmark for designing drugs which can target a subgraph instead of individual proteins.

Journal ArticleDOI
TL;DR: In this paper, a threshold filtering mechanism by processes catalyzed by the enzyme malate dehydrogenase is explained in terms of a model that incorporates an unusual mechanism of inhibition of this enzyme that has a reversible mechanism of action.
Abstract: A recently experimentally observed biochemical "threshold filtering" mechanism by processes catalyzed by the enzyme malate dehydrogenase is explained in terms of a model that incorporates an unusual mechanism of inhibition of this enzyme that has a reversible mechanism of action. Experimental data for a system in which the output signal is produced by biocatalytic processes of the enzyme glucose dehydrogenase are analyzed to verify the model's validity. We also establish that fast reversible conversion of the output product to another compound, without the additional inhibition, cannot on its own result in filtering.

Posted Content
TL;DR: This work considers three representative ERK networks, one with a negative feedback loop, which present a binomial steady state ideal under mass-action kinetics, and applies the theoretical result present in to find a set of rate constants that allow two significantly different stable steady states in the same stoichiometric compatibility class for each network.
Abstract: Mitogen-activated protein kinase (MAPK) signaling pathways play an essential role in the transduction of environmental stimuli to the nucleus, thereby regulating a variety of cellular processes, including cell proliferation, differentiation and programmed cell death. The components of the MAPK extracellular activated protein kinase (ERK) cascade represent attractive targets for cancer therapy as their aberrant activation is a frequent event among highly prevalent human cancers. MAPK networks are a model for computational simulation, mostly using Ordinary and Partial Differential Equations. Key results showed that these networks can have switch-like behavior, bistability and oscillations. In this work, we consider three representative ERK networks, one with a negative feedback loop, which present a binomial steady state ideal under mass-action kinetics. We therefore apply the theoretical result present in P\'erez Mill\'an et. al (2012) to find a set of rate constants that allow two significantly different stable steady states in the same stoichiometric compatibility class for each network. Our approach makes it possible to study certain aspects of the system, such as multistationarity, without relying on simulation, since we do not assume a priori any constant but the topology of the network. As the performed analysis is general it could be applied to many other important biochemical networks.

Journal ArticleDOI
TL;DR: There are close links between these stochastic models and deterministic ODE models of the same networks by embedding the Markov processes into a one-parameter family of processes, where reaction rates and species abundances are scaled in the parameter.
Abstract: We study networks of biochemical reactions modelled by continuous-time Markov processes. Such networks typically contain many molecular species and reactions and are hard to study analytically as well as by simulation. Particularly, we are interested in reaction networks with intermediate species such as the substrate-enzyme complex in the Michaelis-Menten mechanism. These species are virtually in all real-world networks, they are typically short-lived, degraded at a fast rate and hard to observe experimentally. We provide conditions under which the Markov process of a multiscale reaction network with intermediate species is approximated in finite dimensional distribution by the Markov process of a simpler reduced reaction network without intermediate species. We do so by embedding the Markov processes into a one-parameter family of processes, where reaction rates and species abundances are scaled in the parameter. Further, we show that there are close links between these stochastic models and deterministic ODE models of the same networks.

Journal ArticleDOI
TL;DR: This manuscript proposes a practical algorithm for the reduction of biochemical reaction systems using fast-slow asymptotics, which allows the ranking of system variables according to how quickly they approach their momentary steady state, thus selecting the fastest for a steady state approximation.
Abstract: The relationship between components of biochemical network and the resulting dynamics of the overall system is a key focus of computational biology. However, as these networks and resulting mathematical models are inherently complex and non-linear, the understanding of this relationship becomes challenging. Among many approaches, model reduction methods provide an avenue to extract components responsible for the key dynamical features of the system. Unfortunately, these approaches often require intuition to apply. In this manuscript we propose a practical algorithm for the reduction of biochemical reaction systems using fast-slow asymptotics. This method allows the ranking of system variables according to how quickly they approach their momentary steady state, thus selecting the fastest for a steady state approximation. We applied this method to derive models of the Nuclear Factor kappa B network, a key regulator of the immune response that exhibits oscillatory dynamics. Analyses with respect to two specific solutions, which corresponded to different experimental conditions identified different components of the system that were responsible for the respective dynamics. This is an important demonstration of how reduction methods that provide approximations around a specific steady state, could be utilised in order to gain a better understanding of network topology in a broader context.

Posted Content
TL;DR: A strong correlation between relative automorphism group sizes and topological network complexities on the one hand and five year survival probabilities on the other hand is found and this work provides a unifying framework to study protein-protein interaction networks of families of related diseases.
Abstract: Protein-protein interaction networks associated with diseases have gained prominence as an area of research. We investigate algebraic and topological indices for protein-protein interaction networks of 11 human cancers derived from the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. We find a strong correlation between relative automorphism group sizes and topological network complexities on the one hand and five year survival probabilities on the other hand. Moreover, we identify several protein families (e.g. PIK, ITG, AKT families) that are repeated motifs in many of the cancer pathways. Interestingly, these sources of symmetry are often central rather than peripheral. Our results can aide in identification of promising targets for anti-cancer drugs. Beyond that, we provide a unifying framework to study protein-protein interaction networks of families of related diseases (e.g. neurodegenerative diseases, viral diseases, substance abuse disorders).

Journal ArticleDOI
TL;DR: In this article, the authors analyzed the stability of a simple genetic-network model of a living organism under the influence of external and endogenous factors and showed that under most common circumstances a gene network is inherently unstable and suffers from exponential accumulation of gene-regulation deviations leading to death.
Abstract: Several animal species are considered to exhibit what is called negligible senescence, i.e. they do not show signs of functional decline or any increase of mortality with age, and do not have measurable reductions in reproductive capacity with age. Recent studies in Naked Mole Rat (NMR) and long- lived sea urchin showed that the level of gene expression changes with age is lower than in other organisms. These phenotypic observations correlate well with exceptional endurance of NMR tissues to various genotoxic stresses. Therefore, the lifelong transcriptional stability of an organism may be a key determinant of longevity. However, the exact relation between genetic network stability, stress-resistance and aging has not been defined. We analyze the stability of a simple genetic- network model of a living organism under the influence of external and endogenous factors. We demonstrate that under most common circumstances a gene network is inherently unstable and suffers from exponential accumulation of gene-regulation deviations leading to death. However, should the repair systems be sufficiently effective, the gene network can stabilize so that gene damage remains constrained along with mortality of the organism, which may then enjoy a remarkable degree of stability over very long times. We clarify the relation between stress-resistance and aging and suggest that stabilization of the genetic network may provide a mathematical explanation of the Gompertz equation describing the relationship between age and mortality in many species, and of the apparently negligible senescence observed in exceptionally long-lived animals. The model may support a range of applications, such as systematic searches for therapeutics to extend lifespan and healthspan.

Posted Content
TL;DR: This work investigates the effect of realistic extrinsic noises acting on gene deactivation in a common network motif - the positive feedback of a transcription factor on its own synthesis under a variety of settings, i.e., distinct cellular types, distribution of proteins and properties of the external perturbations.
Abstract: It is well-known that gene activation/deactivation dynamics may be a major source of randomness in genetic networks, also in the case of large concentrations of the transcription factors. In this work, we investigate the effect of realistic extrinsic noises acting on gene deactivation in a common network motif - the positive feedback of a transcription factor on its own synthesis - under a variety of settings, i.e., distinct cellular types, distribution of proteins and properties of the external perturbations. At variance with standard models where the perturbations are Gaussian unbounded, we focus here on bounded extrinsic noises to better mimic biological reality. Our results suggest that the gene switching velocity is a key parameter to modulate the response of the network. Simulations suggest that, if the gene switching is fast and many proteins are present, an irreversible noise-induced first order transition is observed as a function of the noise intensity. If noise intensity is further increased a second order transition is also observed. When gene switching is fast, a similar scenario is observed even when few proteins are present, provided that larger cells are considered, which is mainly influenced by noise autocorrelation. On the contrary, if the gene switching is slow, no fist order transitions are observed. In the concluding remarks possible implications of the irreversible transitions for cellular differentiation are briefly discussed.

Journal ArticleDOI
TL;DR: In this paper, the authors outline new directions for conducting cancer systems biology by considering that genome sequencing technology can be used for dissecting, quantifying and genetically characterizing clones from tumors.
Abstract: Recent tumor genome sequencing confirmed that one tumor often consists of multiple cell subpopulations (clones) which bear different, but related, genetic profiles such as mutation and copy number variation profiles. Thus far, one tumor has been viewed as a whole entity in cancer functional studies. With the advances of genome sequencing and computational analysis, we are able to quantify and computationally dissect clones from tumors, and then conduct clone-based analysis. Emerging technologies such as single-cell genome sequencing and RNA-Seq could profile tumor clones. Thus, we should reconsider how to conduct cancer systems biology studies in the genome sequencing era. We will outline new directions for conducting cancer systems biology by considering that genome sequencing technology can be used for dissecting, quantifying and genetically characterizing clones from tumors. Topics discussed in Part 1 of this review include computationally quantifying of tumor subpopulations; clone-based network modeling, cancer hallmark-based networks and their high-order rewiring principles and the principles of cell survival networks of fast-growing clones.

Posted Content
TL;DR: In this article, a distributed classifier is proposed to classify a population of genetically engineered microbial cells by constructing a library of synthetic gene circuits with randomized control sequences (e.g. ribosome-binding sites) in the front element.
Abstract: We describe a conceptual design of a distributed classifier formed by a population of genetically engineered microbial cells. The central idea is to create a complex classifier from a population of weak or simple classifiers. We create a master population of cells with randomized synthetic biosensor circuits that have a broad range of sensitivities towards chemical signals of interest that form the input vectors subject to classification. The randomized sensitivities are achieved by constructing a library of synthetic gene circuits with randomized control sequences (e.g. ribosome-binding sites) in the front element. The training procedure consists in re-shaping of the master population in such a way that it collectively responds to the "positive" patterns of input signals by producing above-threshold output (e.g. fluorescent signal), and below-threshold output in case of the "negative" patterns. The population re-shaping is achieved by presenting sequential examples and pruning the population using either graded selection/counterselection or by fluorescence-activated cell sorting (FACS). We demonstrate the feasibility of experimental implementation of such system computationally using a realistic model of the synthetic sensing gene circuits.

Posted Content
TL;DR: In this paper, the hierarchical structure of the transcriptional regulatory networks (TRNs) of gram-positive bacterium Bacillus subtilis and gram-negative bacterium Escherichia coli was investigated.
Abstract: The transcriptional regulation of gene expression is orchestrated by complex networks of interacting genes. Increasing evidence indicates that these transcriptional regulatory networks (TRNs) in bacteria have an inherently hierarchical architecture, although the design principles and the specific advantages offered by this type of organization have not yet been fully elucidated. In this study, we focussed on the hierarchical structure of the TRN of the gram-positive bacterium Bacillus subtilis and performed a comparative analysis with the TRN of the gram-negative bacterium Escherichia coli. Using a graph-theoretic approach, we organized the transcription factors (TFs) and sigma-factors in the TRNs of B. subtilis and E. coli into three hierarchical levels (Top, Middle and Bottom) and studied several structural and functional properties across them. In addition to many similarities, we found also specific differences, explaining the majority of them with variations in the distribution of sigma-factors across the hierarchical levels in the two organisms. We then investigated the control of target metabolic genes by transcriptional regulators to characterize the differential regulation of three distinct metabolic subsystems (catabolism, anabolism and central energy metabolism). These results suggest that the hierarchical architecture that we observed in B. subtilis represents an effective organization of its TRN to achieve flexibility in the response to diverse stimuli.

Posted Content
TL;DR: This paper shows that constraint-based methods, using reified constraints for expressing the equilibration conditions, make it possible to numerically solve non-linear tropical Equilibration problems, out of reach of standard computation methods.
Abstract: Model reduction is a central topic in systems biology and dynamical systems theory, for reducing the complexity of detailed models, finding important parameters, and developing multi-scale models for instance. While perturbation theory is a standard mathematical tool to analyze the different time scales of a dynamical system, and decompose the system accordingly, tropical methods provide a simple algebraic framework to perform these analyses systematically in polynomial systems. The crux of these tropicalization methods is in the computation of tropical equilibrations. In this paper we show that constraint-based methods, using reified constraints for expressing the equilibration conditions, make it possible to numerically solve non-linear tropical equilibration problems, out of reach of standard computation methods. We illustrate this approach first with the reduction of simple biochemical mechanisms such as the Michaelis-Menten and Goldbeter-Koshland models, and second, with performance figures obtained on a large scale on the model repository \texttt{biomodels.net}

Posted Content
TL;DR: FASTCORMICS outperforms competing algorithms and allows obtaining high-quality, robust models in a high-throughput manner, which will allow the use of metabolic modelling as routine process for the analysis of microarray data e.g. in the field of personalized medicine.
Abstract: Recently we proposed an algorithm for the fast reconstruction of compact context-specific metabolic networks (FASTCORE) that allowed dropping the reconstruction time to the time order of seconds (Vlassis et al.,2014). This extremely low computational demand opens new possibilities for improving the quality of the models. Several rounds of model reconstruction, testing of the model's predictions against real experimental data, curation steps of the input model and the set of core reactions as well as cross-validations assays are required to reconstruct high-quality models. These semi-automated model curations steps are in such extend not possible with competing algorithms due to their high computational demands. To adapt FASTCORE for the integration of microarray data, we therefore propose a new workflow: FASTCORMICS. FASTCORMICS requires as input microarray data and a Genome-scale reconstruction. FASTCORMICS is devoid of heuristic parameter settings and has a low computational demand with overall building times in the order of a few minutes. FASTCORMICS preprocesses the microarrays data with the discretization tool Barcode (Zillox et al, 2007). Barcode uses prior knowledge on the intensity distribution of each probe set for a given microarray platform to segregate between expressed genes and non-expressed genes. This preprocessing step allows circumventing the need of setting a heuristic expression threshold, which is critical for the output models as in response to this threshold alternative pathways or subsystems might be included or excluded, thereby heavily changing the functionalities of the model. In general, FASTCORMICS outperforms competing algorithms and allows obtaining high-quality, robust models in a high-throughput manner. This will allow the use of metabolic modelling as routine process for the analysis of microarray data e.g. in the field of personalized medicine.

Posted Content
TL;DR: In this paper, a graph-theoretic method for conservative biochemical mechanisms characterized by bounded species concentrations is presented, which makes the use of degree theory arguments possible, and illustrates the results with an example of a MAPK network.
Abstract: Biochemical mechanisms with mass action kinetics are often modeled by systems of polynomial differential equations (DE). Determining directly if the DE system has multiple equilibria (multistationarity) is difficult for realistic systems, since they are large, nonlinear and contain many unknown parameters. Mass action biochemical mechanisms can be represented by a directed bipartite graph with species and reaction nodes. Graph-theoretic methods can then be used to assess the potential of a given biochemical mechanism for multistationarity by identifying structures in the bipartite graph referred to as critical fragments. In this article we present a graph-theoretic method for conservative biochemical mechanisms characterized by bounded species concentrations, which makes the use of degree theory arguments possible. We illustrate the results with an example of a MAPK network.

Journal ArticleDOI
TL;DR: Fuzziness (plasticity) of conformational barcodes may not only make the cellular system more plastic, and therefore more adaptable, but may also stabilize the complex system allowing better access to its largest attractor.
Abstract: Conformational barcodes tag functional sites of proteins, and are decoded by interacting molecules transmitting the incoming signal. Conformational barcodes are modified by all co-occurring allosteric events induced by post-translational modifications, pathogen, drug binding, etc. We argue that fuzziness (plasticity) of conformational barcodes may be increased by disordered protein structures, by integrative plasticity of multi-phosphorylation events, by increased intracellular water content (decreased molecular crowding) and by increased action of molecular chaperones. This leads to increased plasticity of signaling and cellular networks. Increased plasticity is both substantiated by and inducing an increased noise level. Using the versatile network dynamics tool, Turbine (this http URL), here we show that the 10% noise level expected in cellular systems shifts a cancer-related signaling network of human cells from its proliferative attractors to its largest, apoptotic attractor representing their health-preserving response in the carcinogen containing and tumor suppressor deficient environment modeled in our study. Thus, fuzzy conformational barcodes may not only make the cellular system more plastic, and therefore more adaptable, but may also stabilize the complex system allowing better access to its largest attractor.

Posted Content
TL;DR: This work considers a canonical biochemical network for receptor readout and maps it onto a typical idealized computational measurement protocol, finding that the biochemical network does not achieve thermodynamic limits of efficiency.
Abstract: Short Abstract — Cells use readout molecules to record the state of cell-surface receptors, apparently analogously to computational measurements. But at what level does this analogy hold? Do living cells operate at the thermodynamic limits of efficiency? We consider a canonical biochemical network for receptor readout and map it onto a typical idealized computational measurement protocol. The biochemical network does not achieve thermodynamic limits of efficiency, facing a tradeoff between dissipation and accuracy of measurement that is more severe than and qualitatively distinct from that required thermodynamically. Biomolecules can, however, perform optimal measurements when the concentrations of ATP and ADP are externally manipulated.

Posted ContentDOI
TL;DR: A large deviation principle is derived so that the QSA can explicitly account for random gene switching without using an adiabatic limit or diffusion approximation, which are unreliable and inaccurate for metastable events.
Abstract: A general class of stochastic gene expression models with self regulation is considered. One or more genes randomly switch between regulatory states, each having a different mRNA transcription rate. The gene or genes are self regulating when the proteins they produce affect the rate of switching between regulatory states. Under weak noise conditions, the deterministic forces are much stronger than fluctuations from gene switching and protein synthesis. Metastable transitions, such as bistable switching, can occur under weak noise conditions, causing dramatic shifts in the expression of a gene. A general tool used to describe metastability is the quasi stationary analysis (QSA). A large deviation principle is derived so that the QSA can explicitly account for random gene switching without using an adiabatic limit or diffusion approximation, which are unreliable and inaccurate for metastable events.This allows the existing asymptotic and numerical methods that have been developed for continuous Markov processes to be used to analyze the full model.

Posted Content
TL;DR: This work proposes a modeling approach derived from Boolean networks where fuzzy operators are used and where edges are tuned, expected to bring enhancements in the ability of qualitative models to simulate the dynamics of biological networks while not requiring quantitative information.
Abstract: Quantitative modeling in systems biology can be difficult due to the scarcity of quantitative details about biological phenomenons, especially at the subcellular scale. An alternative to escape this difficulty is qualitative modeling since it requires few to no quantitative information. Among the qualitative modeling approaches, the Boolean network formalism is one of the most popular. However, Boolean models allow variables to be valued at only true or false, which can appear too simplistic when modeling biological processes. Consequently, this work proposes a modeling approach derived from Boolean networks where fuzzy operators are used and where edges are tuned. Fuzzy operators allow variables to be continuous and then to be more finely valued than with discrete modeling approaches, such as Boolean networks, while remaining qualitative. Moreover, to consider that in a given biological network some interactions are slower and/or weaker relative to other ones, edge states are computed in order to modulate in speed and strength the signal they convey. The proposed formalism is illustrated through its implementation on a tiny sample of the epidermal growth factor receptor signaling pathway. The obtained simulations show that continuous results are produced, thus allowing finer analysis, and that modulating the signal conveyed by the edges allows their tuning according to knowledge about the modeled interactions, thus incorporating more knowledge. The proposed modeling approach is expected to bring enhancements in the ability of qualitative models to simulate the dynamics of biological networks while not requiring quantitative information.

Posted Content
TL;DR: Evaluating MI-GRAAL against a newer approach, GHOST, by mixing-and-matching the methods’ NCFs and ASs to potentially further improve alignment quality and represent general recommendations for a fair evaluation of network alignment methods and in particular of two-stage NCF-AS approaches.
Abstract: Biological network alignment identifies topologically and functionally conserved regions between networks of different species. It encompasses two algorithmic steps: node cost function (NCF), which measures similarities between nodes in different networks, and alignment strategy (AS), which uses these similarities to rapidly identify high-scoring alignments. Different methods use both different NCFs and different ASs. Thus, it is unclear whether the superiority of a method comes from its NCF, its AS, or both. We already showed on MI-GRAAL and IsoRankN that combining NCF of one method and AS of another method can lead to a new superior method. Here, we evaluate MI-GRAAL against newer GHOST to potentially further improve alignment quality. Also, we approach several important questions that have not been asked systematically thus far. First, we ask how much of the node similarity information in NCF should come from sequence data compared to topology data. Existing methods determine this more-less arbitrarily, which could affect the resulting alignment(s). Second, when topology is used in NCF, we ask how large the size of the neighborhoods of the compared nodes should be. Existing methods assume that larger neighborhood sizes are better. We find that MI-GRAAL's NCF is superior to GHOST's NCF, while the performance of the methods' ASs is data-dependent. Thus, the combination of MI-GRAAL's NCF and GHOST's AS could be a new superior method for certain data. Also, which amount of sequence information is used within NCF does not affect alignment quality, while the inclusion of topological information is crucial. Finally, larger neighborhood sizes are preferred, but often, it is the second largest size that is superior, and using this size would decrease computational complexity. Together, our results give several general recommendations for a fair evaluation of network alignment methods.

Posted Content
TL;DR: An automated procedure to determine the relevant positive feedback loops of a multi-stationary reaction network is developed and implemented and useful for interpretation and summary of the network’s dynamics.
Abstract: Bistability is ubiquitous in biological systems. For example, bistability is found in many reaction networks that involve the control and execution of important biological functions, such as signalling processes. Positive feedback loops, composed of species and reactions, are necessary for bistability, and generally for multi-stationarity, to occur. These loops are therefore often used to illustrate and pinpoint the parts of a multi-stationary network that are relevant (`responsible') for the observed multi-stationarity. However positive feedback loops are generally abundant in reaction networks but not all of them are important for subsequent interpretation of the network's dynamics. We present an automated procedure to determine the relevant positive feedback loops of a multi-stationary reaction network. The procedure only reports the loops that are relevant for multi-stationarity (that is, when broken multi-stationarity disappears) and not all positive feedback loops of the network. We show that the relevant positive feedback loops must be understood in the context of the network (one loop might be relevant for one network, but cannot create multi-stationarity in another). Finally, we demonstrate the procedure by applying it to several examples of signaling processes, including a ubiquitination and an apoptosis network, and to models extracted from the Biomodels database. We have developed and implemented an automated procedure to find relevant positive feedback loops in reaction networks. The results of the procedure are useful for interpretation and summary of the network's dynamics.