15 research outputs found

    Genome-scale bacterial transcriptional regulatory networks: reconstruction and integrated analysis with metabolic models

    Get PDF
    Advances in sequencing technology are resulting in the rapid emergence of large numbers of complete genome sequences. High throughput annotation and metabolic modeling of these genomes is now a reality. The high throughput reconstruction and analysis of genome-scale transcriptional regulatory networks represents the next frontier in microbial bioinformatics. The fruition of this next frontier will depend upon the integration of numerous data sources relating to mechanisms, components, and behavior of the transcriptional regulatory machinery, as well as the integration of the regulatory machinery into genome-scale cellular models. Here we review existing repositories for different types of transcriptional regulatory data, including expression data, transcription factor data, and binding site locations, and we explore how these data are being used for the reconstruction of new regulatory networks. From template network based methods to de novo reverse engineering from expression data, we discuss how regulatory networks can be reconstructed and integrated with metabolic models to improve model predictions and performance. Finally, we explore the impact these integrated models can have in simulating phenotypes, optimizing the production of compounds of interest or paving the way to a whole-cell model.J.P.F. acknowledges funding from [SFRH/BD/70824/2010] of the FCT (Portuguese Foundation for Science and Technology) PhD program. The work was supported in part by the ERDF—European Regional Development Fund through the COMPETE Programme (operational programme for competitiveness), National Funds through the FCT within projects [FCOMP-01-0124-FEDER015079] (ToMEGIM—Computational Tools for Metabolic Engineering using Genome-scale Integrated Models) and FCOMP-01-0124-FEDER009707 (HeliSysBio—molecular Systems Biology in Helicobacter pylori), the U.S. Department of Energy under contract [DE-ACO2-06CH11357] and the National Science Foundation under [0850546]

    Netter: re-ranking gene network inference predictions using structural network properties

    Get PDF
    Background: Many algorithms have been developed to infer the topology of gene regulatory networks from gene expression data. These methods typically produce a ranking of links between genes with associated confidence scores, after which a certain threshold is chosen to produce the inferred topology. However, the structural properties of the predicted network do not resemble those typical for a gene regulatory network, as most algorithms only take into account connections found in the data and do not include known graph properties in their inference process. This lowers the prediction accuracy of these methods, limiting their usability in practice. Results: We propose a post-processing algorithm which is applicable to any confidence ranking of regulatory interactions obtained from a network inference method which can use, inter alia, graphlets and several graph-invariant properties to re-rank the links into a more accurate prediction. To demonstrate the potential of our approach, we re-rank predictions of six different state-of-the-art algorithms using three simple network properties as optimization criteria and show that Netter can improve the predictions made on both artificially generated data as well as the DREAM4 and DREAM5 benchmarks. Additionally, the DREAM5 E. coli. community prediction inferred from real expression data is further improved. Furthermore, Netter compares favorably to other post-processing algorithms and is not restricted to correlation-like predictions. Lastly, we demonstrate that the performance increase is robust for a wide range of parameter settings. Netter is available at http://bioinformatics. intec. ugent. be. Conclusions: Network inference from high-throughput data is a long-standing challenge. In this work, we present Netter, which can further refine network predictions based on a set of user-defined graph properties. Netter is a flexible system which can be applied in unison with any method producing a ranking from omics data. It can be tailored to specific prior knowledge by expert users but can also be applied in general uses cases. Concluding, we believe that Netter is an interesting second step in the network inference process to further increase the quality of prediction

    A review of methods for the reconstruction and analysis of integrated genome-scale models of metabolism and regulation

    Get PDF
    The current survey aims to describe the main methodologies for extending the reconstruction and analysis of genome-scale metabolic models and phenotype simulation with Flux Balance Analysis mathematical frameworks, via the integration of Transcriptional Regulatory Networks and/or gene expression data. Although the surveyed methods are aimed at improving phenotype simulations obtained from these models, the perspective of reconstructing integrated genome-scale models of metabolism and gene expression for diverse prokaryotes is still an open challenge.This study was supported by the Portuguese Foundation for Science and Technology (FCT) under the scope of the strategic funding of UIDB/04 469/2020 unit and BioTecNorte operation (NORTE-01-0145-FEDER-000004) funded by the European Regional Development Fund under the scope of Norte2020 -Programa Operacional Regional do Norte. Fernando Cruz holds a doctoral fellowship (SFRH/BD/139198/2018) funded by the FCT. This study was supported by the European Commission through project SHIKIFACTORY100 -Modular cell factories for the production of 100 compounds from the shikimate pathway (Reference 814408). The submitted manuscript has been created by UChicago Argonne, LLC as Operator of Argonne National Laboratory (`Argonne') under Contract No. DE-AC02-06CH11357 with the U.S. Department of Energy. The U.S. Government retains for itself, and others acting on its behalf, a paid-up, nonexclusive, irrevocable worldwide license in said article to reproduce, prepare derivative works, distribute copies to the public, and perform publicly and display publicly, by or on behalf of the Government. The Department of Energy will provide public access to these results of federally sponsored research in accordance with the DOE Public Access Plan.info:eu-repo/semantics/publishedVersio

    Blueprint: descrição da complexidade da regulação metabólica através da reconstrução de modelos metabólicos e regulatórios integrados

    Get PDF
    Tese de doutoramento em Biomedical EngineeringUm modelo metabólico consegue prever o fenótipo de um organismo. No entanto, estes modelos podem obter previsões incorretas, pois alguns processos metabólicos são controlados por mecanismos reguladores. Assim, várias metodologias foram desenvolvidas para melhorar os modelos metabólicos através da integração de redes regulatórias. Todavia, a reconstrução de modelos regulatórios e metabólicos à escala genómica para diversos organismos apresenta diversos desafios. Neste trabalho, propõe-se o desenvolvimento de diversas ferramentas para a reconstrução e análise de modelos metabólicos e regulatórios à escala genómica. Em primeiro lugar, descreve-se o Biological networks constraint-based In Silico Optimization (BioISO), uma nova ferramenta para auxiliar a curação manual de modelos metabólicos. O BioISO usa um algoritmo de relação recursiva para orientar as previsões de fenótipo. Assim, esta ferramenta pode reduzir o número de artefatos em modelos metabólicos, diminuindo a possibilidade de obter erros durante a fase de curação. Na segunda parte deste trabalho, desenvolveu-se um repositório de redes regulatórias para procariontes que permite suportar a sua integração em modelos metabólicos. O Prokaryotic Transcriptional Regulatory Network Database (ProTReND) inclui diversas ferramentas para extrair e processar informação regulatória de recursos externos. Esta ferramenta contém um sistema de integração de dados que converte dados dispersos de regulação em redes regulatórias integradas. Além disso, o ProTReND dispõe de uma aplicação que permite o acesso total aos dados regulatórios. Finalmente, desenvolveu-se uma ferramenta computacional no MEWpy para simular e analisar modelos regulatórios e metabólicos. Esta ferramenta permite ler um modelo metabólico e/ou rede regulatória, em diversos formatos. Esta estrutura consegue construir um modelo regulatório e metabólico integrado usando as interações regulatórias e as ligações entre genes e proteínas codificadas no modelo metabólico e na rede regulatória. Além disso, esta estrutura suporta vários métodos de previsão de fenótipo implementados especificamente para a análise de modelos regulatórios-metabólicos.Genome-Scale Metabolic (GEM) models can predict the phenotypic behavior of organisms. However, these models can lead to incorrect predictions, as certain metabolic processes are controlled by regulatory mechanisms. Accordingly, many methodologies have been developed to extend the reconstruction and analysis of GEM models via the integration of Transcriptional Regulatory Network (TRN)s. Nevertheless, the perspective of reconstructing integrated genome-scale regulatory and metabolic models for diverse prokaryotes is still an open challenge. In this work, we propose several tools to assist the reconstruction and analysis of regulatory and metabolic models. We start by describing BioISO, a novel tool to assist the manual curation of GEM models. BioISO uses a recursive relation-like algorithm and Flux Balance Analysis (FBA) to evaluate and guide debugging of in silico phenotype predictions. Hence, this tool can reduce the number of artifacts in GEM models, decreasing the burdens of model refinement and curation. A state-of-the-art repository of TRNs for prokaryotes was implemented to support the reconstruction and integration of TRNs into GEM models. The ProTReND repository comprehends several tools to extract and process regulatory information available in several resources. More importantly, this repository contains a data integration system to unify the regulatory data into standardized TRNs at the genome scale. In addition, ProTReND contains a web application with full access to the regulatory data. Finally, we have developed a new modeling framework to define, simulate and analyze GEnome-scale Regulatory and Metabolic (GERM) models in MEWpy. The GERM model framework can read a GEM model, as well as a TRN from different file formats. This framework assembles a GERM model using the regulatory interactions and Genes-Proteins-Reactions (GPR) rules encoded into the GEM model and TRN. In addition, this modeling framework supports several methods of phenotype prediction designed for regulatory-metabolic models.I would like to thank Fundação para a Ciência e Tecnologia for the Ph.D. studentship I was awarded with (SFRH/BD/139198/2018)

    Computing and applying atomic regulons to understand gene expression and regulation

    Get PDF
    The Supplementary Material for this article can be found online at: http://journal.frontiersin.org/article/10.3389/fmicb.2016.01819/full#supplementary-materialUnderstanding gene function and regulation is essential for the interpretation prediction and ultimate design of cell responses to changes in the environment. An important step toward meeting the challenge of understanding gene function and regulation is the identification of sets of genes that are always co-expressed. These gene sets Atomic Regulons ARs represent fundamental units of function within a cell and could be used to associate genes of unknown function with cellular processes and to enable rational genetic engineering of cellular systems. Here we describe an approach for inferring ARs that leverages large-scale expression data sets gene context and functional relationships among genes. We computed ARs for Escherichia coli based on 907 gene expression experiments and compared our results with gene clusters produced by two prevalent data-driven methods: hierarchical clustering and k-means clustering. We compared ARs and purely data-driven gene clusters to the curated set of regulatory interactions for E. coli found in RegulonDB showing that ARs are more consistent with gold standard regulons than are data-driven gene clusters. We further examined the consistency of ARs and data-driven gene clusters in the context of gene interactions predicted by Context Likelihood of Relatedness CLR analysis finding that the ARs show better agreement with CLR predicted interactions. We determined the impact of increasing amounts of expression data on AR construction and find that while more data improve ARs it is not necessary to use the full set of gene expression experiments available for E. coli to produce high quality ARs. In order to explore the conservation of co-regulated gene sets across different organisms we computed ARs for Shewanella oneidensis Pseudomonas aeruginosa Thermus thermophilus and Staphylococcus aureus each of which represents increasing degrees of phylogenetic distance from E. coli. Comparison of the organism-specific ARs showed that the consistency of AR gene membership correlates with phylogenetic distance but there is clear variability in the regulatory networks of closely related organisms. As large scale expression data sets become increasingly common for model and non-model organisms comparative analyses of atomic regulons will provide valuable insights into fundamental regulatory modules used across the bacterial domain.JF acknowledges funding from [SFRH/BD/70824/2010] of the FCT (Portuguese Foundation for Science and Technology) PhD program. CH and PW were supported by the National Science Foundation under grant number EFRI-MIKS-1137089. RT was supported by the Genomic Science Program (GSP), Office of Biological and Environmental Research (OBER), U.S. Department of Energy(DOE),and his work is a contribution of the Pacific North west National Laboratory (PNNL) Foundational Scientific Focus Area. This work was partially supported by an award from the National Science Foundation to MD, AB, NT, and RO (NSFABI-0850546). This work was also supported by the United States National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Service [Contract No. HHSN272201400027C]

    Machine learning for biological network inference

    Get PDF

    Plasma levels of interleukin-1 receptor antagonist (IL1Ra) predict radiographic progression of symptomatic knee osteoarthritis

    Get PDF
    Pro- and anti-inflammatory mediators, such as IL-1β and IL1Ra, are produced by joint tissues in osteoarthritis (OA), where they may contribute to pathogenesis. We examined whether inflammatory events occurring within joints are reflected in plasma of patients with symptomatic knee osteoarthritis (SKOA)

    Activity of microRNAs and transcription factors in Gene Regulatory Networks

    Get PDF
    In biological research, diverse high-throughput techniques enable the investigation of whole systems at the molecular level. The development of new methods and algorithms is necessary to analyze and interpret measurements of gene and protein expression and of interactions between genes and proteins. One of the challenges is the integrated analysis of gene expression and the associated regulation mechanisms. The two most important types of regulators, transcription factors (TFs) and microRNAs (miRNAs), often cooperate in complex networks at the transcriptional and post-transcriptional level and, thus, enable a combinatorial and highly complex regulation of cellular processes. For instance, TFs activate and inhibit the expression of other genes including other TFs whereas miRNAs can post-transcriptionally induce the degradation of transcribed RNA and impair the translation of mRNA into proteins. The identification of gene regulatory networks (GRNs) is mandatory in order to understand the underlying control mechanisms. The expression of regulators is itself regulated, i.e. activating or inhibiting regulators in varying conditions and perturbations. Thus, measurements of gene expression following targeted perturbations (knockouts or overexpressions) of these regulators are of particular importance. The prediction of the activity states of the regulators and the prediction of the target genes are first important steps towards the construction of GRNs. This thesis deals with these first bioinformatics steps to construct GRNs. Targets of TFs and miRNAs are determined as comprehensively and accurately as possible. The activity state of regulators is predicted for specific high-throughput data and specific contexts using appropriate statistical approaches. Moreover, (parts of) GRNs are inferred, which lead to explanations of given measurements. The thesis describes new approaches for these tasks together with accompanying evaluations and validations. This immediately defines the three main goals of the current thesis: 1. The development of a comprehensive database of regulator-target relation. Regulators and targets are retrieved from public repositories, extracted from the literature via text mining and collected into the miRSel database. In addition, relations can be predicted using various published methods. In order to determine the activity states of regulators (see 2.) and to infer GRNs (3.) comprehensive and accurate regulator-target relations are required. It could be shown that text mining enables the reliable extraction of miRNA, gene, and protein names as well as their relations from scientific free texts. Overall, the miRSel contains about three times more relations for the model organisms human, mouse, and rat as compared to state-of-the-art databases (e.g. TarBase, one of the currently most used resources for miRNA-target relations). 2. The prediction of activity states of regulators based on improved target sets. In order to investigate mechanisms of gene regulation, the experimental contexts have to be determined in which the respective regulators become active. A regulator is predicted as active based on appropriate statistical tests applied to the expression values of its set of target genes. For this task various gene set enrichment (GSE) methods have been proposed. Unfortunately, before an actual experiment it is unknown which genes are affected. The missing standard-of-truth so far has prevented the systematic assessment and evaluation of GSE tests. In contrast, the trigger of gene expression changes is of course known for experiments where a particular regulator has been directly perturbed (i.e. by knockout, transfection, or overexpression). Based on such datasets, we have systematically evaluated 12 current GSE tests. In our analysis ANOVA and the Wilcoxon test performed best. 3. The prediction of regulation cascades. Using gene expression measurements and given regulator-target relations (e.g. from the miRSel database) GRNs are derived. GSE tests are applied to determine TFs and miRNAs that change their activity as cellular response to an overexpressed miRNA. Gene regulatory networks can constructed iteratively. Our models show how miRNAs trigger gene expression changes: either directly or indirectly via cascades of miRNA-TF, miRNA-kinase-TF as well as TF-TF relations. In this thesis we focus on measurements which have been obtained after overexpression of miRNAs. Surprisingly, a number of cancer relevant miRNAs influence a common core of TFs which are involved in processes such as proliferation and apoptosis

    Network-based analysis of gene expression data

    Get PDF
    The methods of molecular biology for the quantitative measurement of gene expression have undergone a rapid development in the past two decades. High-throughput assays with the microarray and RNA-seq technology now enable whole-genome studies in which several thousands of genes can be measured at a time. However, this has also imposed serious challenges on data storage and analysis, which are subject of the young, but rapidly developing field of computational biology. To explain observations made on such a large scale requires suitable and accordingly scaled models of gene regulation. Detailed models, as available for single genes, need to be extended and assembled in larger networks of regulatory interactions between genes and gene products. Incorporation of such networks into methods for data analysis is crucial to identify molecular mechanisms that are drivers of the observed expression. As methods for this purpose emerge in parallel to each other and without knowing the standard of truth, results need to be critically checked in a competitive setup and in the context of the available rich literature corpus. This work is centered on and contributes to the following subjects, each of which represents important and distinct research topics in the field of computational biology: (i) construction of realistic gene regulatory network models; (ii) detection of subnetworks that are significantly altered in the data under investigation; and (iii) systematic biological interpretation of detected subnetworks. For the construction of regulatory networks, I review existing methods with a focus on curation and inference approaches. I first describe how literature curation can be used to construct a regulatory network for a specific process, using the well-studied diauxic shift in yeast as an example. In particular, I address the question how a detailed understanding, as available for the regulation of single genes, can be scaled-up to the level of larger systems. I subsequently inspect methods for large-scale network inference showing that they are significantly skewed towards master regulators. A recalibration strategy is introduced and applied, yielding an improved genome-wide regulatory network for yeast. To detect significantly altered subnetworks, I introduce GGEA as a method for network-based enrichment analysis. The key idea is to score regulatory interactions within functional gene sets for consistency with the observed expression. Compared to other recently published methods, GGEA yields results that consistently and coherently align expression changes with known regulation types and that are thus easier to explain. I also suggest and discuss several significant enhancements to the original method that are improving its applicability, outcome and runtime. For the systematic detection and interpretation of subnetworks, I have developed the EnrichmentBrowser software package. It implements several state-of-the-art methods besides GGEA, and allows to combine and explore results across methods. As part of the Bioconductor repository, the package provides a unified access to the different methods and, thus, greatly simplifies the usage for biologists. Extensions to this framework, that support automating of biological interpretation routines, are also presented. In conclusion, this work contributes substantially to the research field of network-based analysis of gene expression data with respect to regulatory network construction, subnetwork detection, and their biological interpretation. This also includes recent developments as well as areas of ongoing research, which are discussed in the context of current and future questions arising from the new generation of genomic data
    corecore