71 research outputs found

    Framework for network modularization and Bayesian network analysis to investigate the perturbed metabolic network

    Get PDF
    Background: Genome-scale metabolic network models have contributed to elucidating biological phenomena, and predicting gene targets to engineer for biotechnological applications. With their increasing importance, their precise network characterization has also been crucial for better understanding of the cellular physiology.Results: We herein introduce a framework for network modularization and Bayesian network analysis (FMB) to investigate organism's metabolism under perturbation. FMB reveals direction of influences among metabolic modules, in which reactions with similar or positively correlated flux variation patterns are clustered, in response to specific perturbation using metabolic flux data. With metabolic flux data calculated by constraints-based flux analysis under both control and perturbation conditions, FMB, in essence, reveals the effects of specific perturbations on the biological system through network modularization and Bayesian network analysis at metabolic modular level. As a demonstration, this framework was applied to the genetically perturbed Escherichia coli metabolism, which is a lpdA gene knockout mutant, using its genome-scale metabolic network model.Conclusions: After all, it provides alternative scenarios of metabolic flux distributions in response to the perturbation, which are complementary to the data obtained from conventionally available genome-wide high-throughput techniques or metabolic flux analysis

    Mathematical models of cellular decisions: investigating immune response and apoptosis

    No full text
    The main objective of this thesis is to develop and analyze mathematical models of cellular decisions. This work focuses on understanding the mechanisms involved in specific cellular processes such as immune response in the vascular system, and those involved in apoptosis, or programmed cellular death. A series of simple ordinary differential equation (ODE) models are constructed describing the macrophage response to hemoglobin:haptoglobin (Hb:Hp) complexes that may be present in vascular inflammation. The models proposed a positive feedback loop between the CD163 macrophage receptor and anti-inflammatory cytokine interleukin-10 (IL-10) and bifurcation analysis predicted the existence of a cellular phenotypic switch which was experimentally verified. Moreover, these models are extended to include the intracellular mediator heme oxygenase-1 (HO-1). Analysis of the proposed models find a positive feedback mechanism between IL-10 and HO-1. This model also predicts cellular response of heme and IL-10 stimuli. For the apoptotic (cell suicide) system, a modularized model is constructed encompassing the extrinsic and intrinsic signaling pathways. Model reduction is performed by abstracting the dynamics of complexes (oligomers) at a steady-state. This simplified model is analyzed, revealing different kinetic properties between type I and type II cells, and reduced models verify results. The second model of apoptosis proposes a novel mechanism of apoptosis activation through receptor-ligand clustering, yielding robust bistability and hysteresis. Using techniques from algebraic geometry, a model selection criterion is provided between the proposed and existing model as experimental data becomes available to verify the mechanism. The models developed throughout this thesis reveal important and relevant mechanisms specific to cellular response; specifically, interactions necessary for an organism to maintain homeostasis are identified. This work enables a deeper understanding of the biological interactions and dynamics of vascular inflammation and apoptosis. The results of these models provide predictions which may motivate further experimental work and theoretical study

    Computational Labeling, Partitioning, and Balancing of Molecular Networks

    Get PDF
    Recent advances in high throughput techniques enable large-scale molecular quantification with high accuracy, including mRNAs, proteins and metabolites. Differential expression of these molecules in case and control samples provides a way to select phenotype-associated molecules with statistically significant changes. However, given the significance ranking list of molecular changes, how those molecules work together to drive phenotype formation is still unclear. In particular, the changes in molecular quantities are insufficient to interpret the changes in their functional behavior. My study is aimed at answering this question by integrating molecular network data to systematically model and estimate the changes of molecular functional behaviors. We build three computational models to label, partition, and balance molecular networks using modern machine learning techniques. (1) Due to the incompleteness of protein functional annotation, we develop AptRank, an adaptive PageRank model for protein function prediction on bilayer networks. By integrating Gene Ontology (GO) hierarchy with protein-protein interaction network, our AptRank outperforms four state-of-the-art methods in a comprehensive evaluation using benchmark datasets. (2) We next extend our AptRank into a network partitioning method, BioSweeper, to identify functional network modules in which molecules share similar functions and also densely connect to each other. Compared to traditional network partitioning methods using only network connections, BioSweeper, which integrates the GO hierarchy, can automatically identify functionally enriched network modules. (3) Finally, we conduct a differential interaction analysis, namely difFBA, on protein-protein interaction networks by simulating protein fluxes using flux balance analysis (FBA). We test difFBA using quantitative proteomic data from colon cancer, and demonstrate that difFBA offers more insights into functional changes in molecular behavior than does protein quantity changes alone. We conclude that our integrative network model increases the observational dimensions of complex biological systems, and enables us to more deeply understand the causal relationships between genotypes and phenotypes

    Role of network topology based methods in discovering novel gene-phenotype associations

    Get PDF
    The cell is governed by the complex interactions among various types of biomolecules. Coupled with environmental factors, variations in DNA can cause alterations in normal gene function and lead to a disease condition. Often, such disease phenotypes involve coordinated dysregulation of multiple genes that implicate inter-connected pathways. Towards a better understanding and characterization of mechanisms underlying human diseases, here, I present GUILD, a network-based disease-gene prioritization framework. GUILD associates genes with diseases using the global topology of the protein-protein interaction network and an initial set of genes known to be implicated in the disease. Furthermore, I investigate the mechanistic relationships between disease-genes and explain the robustness emerging from these relationships. I also introduce GUILDify, an online and user-friendly tool which prioritizes genes for their association to any user-provided phenotype. Finally, I describe current state-of-the-art systems-biology approaches where network modeling has helped extending our view on diseases such as cancer.La cèl•lula es regeix per interaccions complexes entre diferents tipus de biomolècules. Juntament amb factors ambientals, variacions en el DNA poden causar alteracions en la funció normal dels gens i provocar malalties. Sovint, aquests fenotips de malaltia involucren una desregulació coordinada de múltiples gens implicats en vies interconnectades. Per tal de comprendre i caracteritzar millor els mecanismes subjacents en malalties humanes, en aquesta tesis presento el programa GUILD, una plataforma que prioritza gens relacionats amb una malaltia en concret fent us de la topologia de xarxe. A partir d’un conjunt conegut de gens implicats en una malaltia, GUILD associa altres gens amb la malaltia mitjancant la topologia global de la xarxa d’interaccions de proteïnes. A més a més, analitzo les relacions mecanístiques entre gens associats a malalties i explico la robustesa es desprèn d’aquesta anàlisi. També presento GUILDify, un servidor web de fácil ús per la priorització de gens i la seva associació a un determinat fenotip. Finalment, descric els mètodes més recents en què el model•latge de xarxes ha ajudat extendre el coneixement sobre malalties complexes, com per exemple a càncer

    Novel modeling formalisms and simulation tools in computational biosystems

    Get PDF
    Tese de doutoramento em BioengenhariaThe goal of Systems Biology is to understand the complex behavior that emerges from the interaction among the cellular components. Industrial biotechnology is one of the areas of application, where new approaches for metabolic engineering are developed, through the creation of new models and tools for simulation and optimization of the microbial metabolism. Although whole-cell modeling is one of the goals of Systems Biology, so far most models address only one kind of biological network independently. This work explores the integration of di erent kinds of biological networks with a focus on the improvement of simulation of cellular metabolism. The bacterium Escherichia coli is the most well characterized model organism and is used as our case-study. An extensive review of modeling formalisms that have been used in Systems Biology is presented in this work. It includes several formalisms, including Boolean networks, Bayesian networks, Petri nets, process algebras, constraint-based models, di erential equations, rule-based models, interacting state machines, cellular automata and agent-based models. We compare the features provided by these formalisms and classify the most suitable ones for the creation of a common framework for modeling, analysis and simulation of integrated biological networks. Currently, there is a separation between dynamic and constraint-based modeling of metabolism. Dynamic models are based on detailed kinetic reconstructions of central metabolic pathways, whereas constraint-based models are based on genome-scale stoichiometric reconstructions. Here, we explore the gap between both formulations and evaluate how dynamic models can be used to reduce the solution space of constraint-based models in order to eliminate kinetically infeasible solutions. The limitations of both kinds of models are leading to new approaches to build kinetic models at the genome-scale. The generation of kinetic models from stoichiometric reconstructions can be performed within the same framework as a transformation from discrete to continuous Petri nets. However, the size of these networks results in models with a large number of parameters. In this scope, we develop and implement structural reduction methods that adjust the level of detail of metabolic networks without loss of information, which can be applied prior to the kinetic inference to build dynamic models with a smaller number of parameters. In order to account for enzymatic regulation, which is not present in constraint-based models, we propose the utilization of Extended Petri nets. This results in a better sca old for the kinetic inference process. We evaluate the impact of accounting for enzymatic regulation in the simulation of the steady-state phenotype of mutant strains by performing knockouts and adjustment of enzyme expression levels. It can be observed that in some cases the impact is signi cant and may reveal new targets for rational strain design. In summary, we have created a solid framework with a common formalism and methods for metabolic modeling. This will facilitate the integration with gene regulatory networks, as we have already addressed many issues also associated with these networks, such as the trade-o between size and detail, and the representation of regulatory interactions.O objectivo da Biologia de Sistemas é compreender os comportamentos que resultam das complexas interacções entre todos os componentes celulares. A biotecnologia industrial é uma das áreas de aplicação, onde novas abordagens para a engenharia metabólica são desenvolvidas através da criação de novos modelos e ferramentas de simulação e optimização do metabolismo microbiano. Apesar de um dos principais objectivos da Biologia de Sistemas ser a criação de um modelo completo de uma célula, até ao momento a maioria dos modelos desenvolvidos incorpora de forma separada cada tipo de rede biológica. Este trabalho explora a integração de diferentes tipos de redes biológicas, focando melhorar a simulação do metabolismo celular. A bactéria Escherichia coli é o organismo modelo que estáa melhor caracterizado e é usado como caso de estudo. Neste trabalho é elaborada uma extensa revisão dos formalismos de modela ção que têm sido utilizados em Biologia de Sistemas. São considerados vários formalismos tais como, redes Booleanas, redes Bayesianas, redes de Petri, álgebras de processos, modelos baseados em restrições, equações diferenciais, modelos baseados em regras, máquinas de interacção de estados, autómatos celulares e modelos baseados em agentes. As funcionalidades inerentes a estes formalismos são analisadas de forma a classificar os mesmos pelo seu potencial em servir de base à criação de uma plataforma para modela ção, análise e simulação de redes biológicas integradas. Actualmente, existe uma separação entre modelação dinâmica e modelação baseada em restrições para o metabolismo celular. Os modelos dinâmicos consistem em reconstruções cinéticas detalhadas de vias centrais do metabolismo, enquanto que os modelos baseados em restrições são construídos à escala genómica com base apenas na estequiometria das reacçõoes. Neste trabalho explora-se a separação entre os dois tipos de formulação e é avaliada a forma como os modelos dinâmicos podem ser utilizados para reduzir o espaço de soluções de modelos baseados em restrições de forma a eliminar soluções inalcançáveis. As limitações impostas por ambos os tipos de modelos estão a conduzir à criação de novas abordagens para a construção de modelos cinéticos à escala genómica. A geração de modelos cinéticos a partir de reconstruções estequiométricas pode ser feita dentro de um mesmo formalismo através da transformação de redes de Petri discretas em redes de Petri contínuas. No entanto, devido ao tamanho destas redes, os modelos resultantes incluem um número extremamente grande de parâmetros. Neste trabalho são implementados métodos para a redução estrutural de redes metabólicas sem perda de informação, que permitem ajustar o nível de detalhe das redes. Estes métodos podem ser aplicados à inferência de cinéticas, de forma a gerar modelos dinâmicos com um menor número de parâmetros. De forma a considerar efeitos de regulação enzimática, os quais não são representados em modelos baseados em restrições, propõe-se a utilização de redes de Petri complementadas com arcos regulatórios. Este formalismo é utilizado como base para o processo de inferência cinética. A influência da regulação enzimática na determinação do estado estacionário de estirpes mutantes é avaliada através da análise da remoção de reacções e da variação dos níveis de expressão enzimática. Observa-se que em alguns casos esta influência é significativa e pode ser utilizada para obter novas estratégias de manipulação de estirpes. Em suma, neste trabalho foi criada uma plataforma sólida para modelação do metabolismo baseada num formalismo comum. Esta plataforma facilitará a integração com redes de regulação genética, pois foram abordados vários problemas que também se colocam nestas redes, tais como o ajuste entre o tamanho da rede e o seu nível de detalhe, bem como a representação de interacções regulatórias entre componentes da rede

    Metabolomic Analysis of Defense-Related Reprogramming in Sorghum bicolor in Response to Colletotrichum sublineolum Infection Reveals a Functional Metabolic Web of Phenylpropanoid and Flavonoid Pathways

    Get PDF
    The metabolome of a biological system provides a functional readout of the cellular state, thus serving as direct signatures of biochemical events that define the dynamic equilibrium of metabolism and the correlated phenotype. Hence, to elucidate biochemical processes involved in sorghum responses to fungal infection, a liquid chromatography-mass spectrometry-based untargeted metabolomic study was designed. Metabolic alterations of three sorghum cultivars responding to Colletotrichum sublineolum, were investigated. At the 4-leaf growth stage, the plants were inoculated with fungal spore suspensions and the infection monitored over time: 0, 3, 5, 7, and 9 days post inoculation. Non-infected plants were used as negative controls. The metabolite composition of aqueous-methanol extracts were analyzed on an ultra-high performance liquid chromatography system coupled to high-definition mass spectrometry. The acquired multidimensional data were processed to create data matrices for multivariate statistical analysis and chemometric modeling. The computed chemometric models indicated time- and cultivar-related metabolic changes that reflect sorghum responses to the fungal infection. Metabolic pathway and correlation-based network analyses revealed that this multi-component defense response is characterized by a functional metabolic web, containing defense-related molecular cues to counterattack the pathogen invasion. Components of this network are metabolites from a range of interconnected metabolic pathways with the phenylpropanoid and flavonoid pathways being the central hub of the web. One of the key features of this altered metabolism was the accumulation of an array of phenolic compounds, particularly de novo biosynthesis of the antifungal 3-deoxyanthocynidin phytoalexins, apigeninidin, luteolinidin, and related conjugates. The metabolic results were complemented by qRT-PCR gene expression analyses that showed upregulation of defense-related marker genes. Unraveling key characteristics of the biochemical mechanism underlying sorghum—C. sublineolum interactions, provided valuable insights with potential applications in breeding crop plants with enhanced disease resistance. Furthermore, the study contributes to ongoing efforts toward a comprehensive understanding of the regulation and reprogramming of plant metabolism under biotic stress

    A Network Biology Approach Identifies Molecular Cross-Talk between Normal Prostate Epithelial and Prostate Carcinoma Cells

    Get PDF
    © 2016 The Authors. Published by Public Library of Science. This is an open access article available under a Creative Commons licence. The published version can be accessed at the following link on the publisher’s website: https://doi.org/10.1371/journal.pcbi.1004884The advent of functional genomics has enabled the genome-wide characterization of the molecular state of cells and tissues, virtually at every level of biological organization. The difficulty in organizing and mining this unprecedented amount of information has stimulated the development of computational methods designed to infer the underlying structure of regulatory networks from observational data. These important developments had a profound impact in biological sciences since they triggered the development of a novel data-driven investigative approach. In cancer research, this strategy has been particularly successful. It has contributed to the identification of novel biomarkers, to a better characterization of disease heterogeneity and to a more in depth understanding of cancer pathophysiology. However, so far these approaches have not explicitly addressed the challenge of identifying networks representing the interaction of different cell types in a complex tissue. Since these interactions represent an essential part of the biology of both diseased and healthy tissues, it is of paramount importance that this challenge is addressed. Here we report the definition of a network reverse engineering strategy designed to infer directional signals linking adjacent cell types within a complex tissue. The application of this inference strategy to prostate cancer genome-wide expression profiling data validated the approach and revealed that normal epithelial cells exert an anti-tumour activity on prostate carcinoma cells. Moreover, by using a Bayesian hierarchical model integrating genetics and gene expression data and combining this with survival analysis, we show that the expression of putative cell communication genes related to focal adhesion and secretion is affected by epistatic gene copy number variation and it is predictive of patient survival. Ultimately, this study represents a generalizable approach to the challenge of deciphering cell communication networks in a wide spectrum of biological systems.Cancer research UK, BBSRC, NI

    Network architecture for prediction of emergence in complex biological systems

    Get PDF
    Emergence of properties at the system level, where these properties are not observed at the individual entity level, is an important feature of complex systems. Biological system emergent properties have critical roles in the functioning of organisms and the disruptions to normal functioning, and are relevant to the treatment of diseases like cancer. Complex biological systems can be modeled by abstractions in the form of molecular networks like gene regulatory networks (GRNs) and signaling networks with nodes representing molecules like genes and edges representing molecular interactions. The thesis aims at exploring the use of the architecture of these networks to predict emergence of system properties. First, to better infer the network architecture with aspects that can be useful in predicting emergence, we propose a novel algorithm Polynomial Lasso Bagging or PoLoBag for signed GRN inference from gene expression data. The GRN edge signs represent the nature of the regulatory relationships, activating or inhibitory. Our algorithm gives more accurate signed inference compared to state-of-the-art algorithms, and overcomes their weaknesses by also inferring edge directions and cycles. We also show how combining signed GRN architecture with dynamical information in our proposed dynamical K-core method predicts emergent states of the gene regulatory system effectively. Second, we investigate the existence of the bow-tie architectural organization in the GRNs of species of widely varying complexity. Prior work has shown the existence of this bow-tie feature in the GRNs of only some eukaryotes. Our investigation covers GRNs of prokaryotes to unicellular and multicellular eukaryotes. We find that the observed bow-tie architecture is a characteristic feature of GRNs. Based on differences that we observe in the bow-tie architectures across species, we predict a trend in the emergence of the dynamical gene regulatory system property of controllability with varying species complexity. Third, from input genotype data we predict an emergent phenotype at the organism level -- the cancer-specific survival risk. We propose a novel Mutated Pathway Visible Neural Network or MPVNN, designed using prior knowledge of signaling network architecture and additional mutation data-based edge randomization. This randomization models how known signaling network architecture changes for a particular cancer type, which is not modeled by state-of-the-art visible neural networks. We suggest that MPVNN performs cancer-specific risk prediction better than other similar sized NN and non-NN survival analysis methods, while also providing reliable interpretations of the predictions. These three research contributions taken together make significant advances towards our goal of using molecular network architecture for better prediction of emergence, which can inform treatment decisions and lead to novel therapeutic approaches and is of value to computational biologists and clinicians

    Moving Towards Predictive Toxicology - A Systems Biology Approach

    Get PDF
    Human health and the environment are at risk of being exposed to a large number of hazardous chemicals each day. Unfortunately, many of these chemicals have no or little recorded toxicity information. Predictive toxicology aims to provide tools and methodologies to address this issue. In combination with systems biology approaches these can provide a powerful toolbox for understanding the impact of chemicals on biological species. The work presented within this thesis demonstrates the e ectiveness of such approaches in the context of industrial and environmentally relevant species. More specifically we focus on characterization of a general toxicity mechanism in Rattus norvegius and Daphnia magna as well as for the first time demonstrate that the transcriptional response of D. magna is predictive not only of chemical class but also of measured toxicity. We also show that inclusion of pathwaylevel information can increase biological interpretability in non-model species. Lastly, we provide evidence supporting the application of reverse engineering methodologies in the context of identifying adverse outcome pathways in Pimephales promelas, an environmentally relevant species. Ultimately, our results have shown that these approaches can provide highly relevant information in model and non-model species. Further development building on these results could potentially lead to improvements in risk assessment and environmental monitoring
    corecore