244
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Computational prediction of molecular pathogen-host interactions based on dual transcriptome data

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Inference of inter-species gene regulatory networks based on gene expression data is an important computational method to predict pathogen-host interactions (PHIs). Both the experimental setup and the nature of PHIs exhibit certain characteristics. First, besides an environmental change, the battle between pathogen and host leads to a constantly changing environment and thus complex gene expression patterns. Second, there might be a delay until one of the organisms reacts. Third, toward later time points only one organism may survive leading to missing gene expression data of the other organism. Here, we account for PHI characteristics by extending NetGenerator, a network inference tool that predicts gene regulatory networks from gene expression time series data. We tested multiple modeling scenarios regarding the stimuli functions of the interaction network based on a benchmark example. We show that modeling perturbation of a PHI network by multiple stimuli better represents the underlying biological phenomena. Furthermore, we utilized the benchmark example to test the influence of missing data points on the inference performance. Our results suggest that PHI network inference with missing data is possible, but we recommend to provide complete time series data. Finally, we extended the NetGenerator tool to incorporate gene- and time point specific variances, because complex PHIs may lead to high variance in expression data. Sample variances are directly considered in the objective function of NetGenerator and indirectly by testing the robustness of interactions based on variance dependent disturbance of gene expression values. We evaluated the method of variance incorporation on dual RNA sequencing (RNA-Seq) data of Mus musculus dendritic cells incubated with Candida albicans and proofed our method by predicting previously verified PHIs as robust interactions.

          Related collections

          Most cited references38

          • Record: found
          • Abstract: found
          • Article: not found

          The transcriptional landscape of the yeast genome defined by RNA sequencing.

          The identification of untranslated regions, introns, and coding regions within an organism remains challenging. We developed a quantitative sequencing-based method called RNA-Seq for mapping transcribed regions, in which complementary DNA fragments are subjected to high-throughput sequencing and mapped to the genome. We applied RNA-Seq to generate a high-resolution transcriptome map of the yeast genome and demonstrated that most (74.5%) of the nonrepetitive sequence of the yeast genome is transcribed. We confirmed many known and predicted introns and demonstrated that others are not actively used. Alternative initiation codons and upstream open reading frames also were identified for many yeast genes. We also found unexpected 3'-end heterogeneity and the presence of many overlapping genes. These results indicate that the yeast transcriptome is more complex than previously appreciated.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            featureCounts: An efficient general-purpose program for assigning sequence reads to genomic features

            , , (2013)
            Next-generation sequencing technologies generate millions of short sequence reads, which are usually aligned to a reference genome. In many applications, the key information required for downstream analysis is the number of reads mapping to each genomic feature, for example to each exon or each gene. The process of counting reads is called read summarization. Read summarization is required for a great variety of genomic analyses but has so far received relatively little attention in the literature. We present featureCounts, a read summarization program suitable for counting reads generated from either RNA or genomic DNA sequencing experiments. featureCounts implements highly efficient chromosome hashing and feature blocking techniques. It is considerably faster than existing methods (by an order of magnitude for gene-level summarization) and requires far less computer memory. It works with either single or paired-end reads and provides a wide range of options appropriate for different sequencing applications. featureCounts is available under GNU General Public License as part of the Subread (http://subread.sourceforge.net) or Rsubread (http://www.bioconductor.org) software packages.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Gene regulatory network inference: data integration in dynamic models-a review.

              Systems biology aims to develop mathematical models of biological systems by integrating experimental and theoretical techniques. During the last decade, many systems biological approaches that base on genome-wide data have been developed to unravel the complexity of gene regulation. This review deals with the reconstruction of gene regulatory networks (GRNs) from experimental data through computational methods. Standard GRN inference methods primarily use gene expression data derived from microarrays. However, the incorporation of additional information from heterogeneous data sources, e.g. genome sequence and protein-DNA interaction data, clearly supports the network inference process. This review focuses on promising modelling approaches that use such diverse types of molecular biological information. In particular, approaches are discussed that enable the modelling of the dynamics of gene regulatory systems. The review provides an overview of common modelling schemes and learning algorithms and outlines current challenges in GRN modelling.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Microbiol
                Front Microbiol
                Front. Microbiol.
                Frontiers in Microbiology
                Frontiers Media S.A.
                1664-302X
                06 February 2015
                2015
                : 6
                : 65
                Affiliations
                [1] 1Department of Systems Biology and Bioinformatics, Leibniz-Institute for Natural Product Research and Infection Biology – Hans-Knoell-Institute Jena, Germany
                [2] 2BioControl Jena GmbH Jena, Germany
                Author notes

                Edited by: Saliha Durmus, Gebze Technical University, Turkey

                Reviewed by: Ikbal Agah Ince, Wageningen University and Research Centrum, Netherlands; Kazim Yalcin Arga, Marmara University, Turkey

                *Correspondence: Jörg Linde, Department of Systems Biology and Bioinformatics, Leibniz-Institute for Natural Product Research and Infection Biology – Hans-Knoell-Institute, Beutenbergstr. 11a, 07745 Jena, Germany e-mail: joerg.linde@ 123456hki-jena.de

                This article was submitted to Infectious Diseases, a section of the journal Frontiers in Microbiology.

                Article
                10.3389/fmicb.2015.00065
                4319478
                3239f6d6-aa55-4256-b74a-28e7975b8bd4
                Copyright © 2015 Schulze, Henkel, Driesch, Guthke and Linde.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 11 December 2014
                : 19 January 2015
                Page count
                Figures: 3, Tables: 0, Equations: 8, References: 57, Pages: 11, Words: 8413
                Categories
                Public Health
                Original Research Article

                Microbiology & Virology
                network inference,netgenerator,transcriptomics,dual rna-seq,microarrays,gene regulatory networks,inter-species interactions

                Comments

                Comment on this article