1051
|
Arighi C, Shamovsky V, Masci AM, Ruttenberg A, Smith B, Natale DA, Wu C, D’Eustachio P. Toll-like receptor signaling in vertebrates: testing the integration of protein, complex, and pathway data in the protein ontology framework. PLoS One 2015; 10:e0122978. [PMID: 25894391 PMCID: PMC4404318 DOI: 10.1371/journal.pone.0122978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 02/26/2015] [Indexed: 11/20/2022] Open
Abstract
The Protein Ontology (PRO) provides terms for and supports annotation of species-specific protein complexes in an ontology framework that relates them both to their components and to species-independent families of complexes. Comprehensive curation of experimentally known forms and annotations thereof is expected to expose discrepancies, differences, and gaps in our knowledge. We have annotated the early events of innate immune signaling mediated by Toll-Like Receptor 3 and 4 complexes in human, mouse, and chicken. The resulting ontology and annotation data set has allowed us to identify species-specific gaps in experimental data and possible functional differences between species, and to employ inferred structural and functional relationships to suggest plausible resolutions of these discrepancies and gaps.
Collapse
Affiliation(s)
- Cecilia Arighi
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, Delaware, United States of America
| | - Veronica Shamovsky
- Department of Biochemistry & Molecular Pharmacology, NYU School of Medicine, New York, New York, United States of America
| | - Anna Maria Masci
- Department of Immunology, Duke University, Durham, North Carolina, United States of America
| | - Alan Ruttenberg
- School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Barry Smith
- Department of Philosophy and Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Darren A. Natale
- Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D. C., United States of America
| | - Cathy Wu
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, Delaware, United States of America
- Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D. C., United States of America
| | - Peter D’Eustachio
- Department of Biochemistry & Molecular Pharmacology, NYU School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
1052
|
Groza T, Tudorache T, Robinson PN, Zankl A. Capturing domain knowledge from multiple sources: the rare bone disorders use case. J Biomed Semantics 2015; 6:21. [PMID: 25926964 PMCID: PMC4414390 DOI: 10.1186/s13326-015-0008-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 03/02/2015] [Indexed: 12/13/2022] Open
Abstract
Background Lately, ontologies have become a fundamental building block in the process of formalising and storing complex biomedical information. The community-driven ontology curation process, however, ignores the possibility of multiple communities building, in parallel, conceptualisations of the same domain, and thus providing slightly different perspectives on the same knowledge. The individual nature of this effort leads to the need of a mechanism to enable us to create an overarching and comprehensive overview of the different perspectives on the domain knowledge. Results We introduce an approach that enables the loose integration of knowledge emerging from diverse sources under a single coherent interoperable resource. To accurately track the original knowledge statements, we record the provenance at very granular levels. We exemplify the approach in the rare bone disorders domain by proposing the Rare Bone Disorders Ontology (RBDO). Using RBDO, researchers are able to answer queries, such as: “What phenotypes describe a particular disorder and are common to all sources?” or to understand similarities between disorders based on divergent groupings (classifications) provided by the underlying sources. Availability RBDO is available at http://purl.org/skeletome/rbdo. In order to support lightweight query and integration, the knowledge captured by RBDO has also been made available as a SPARQL Endpoint at http://bio-lark.org/se_skeldys.html.
Collapse
Affiliation(s)
- Tudor Groza
- School of ITEE, The University of Queensland, St Lucia, Australia
| | - Tania Tudorache
- Stanford Center for Biomedical Informatics Research, Stanford University, Stanford, USA
| | - Peter N Robinson
- Institut für Medizinische Genetik und Humangenetik, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Zankl
- Children's Hospital, Westmead, The University of Sydney, Sydney, New South Wales Australia
| |
Collapse
|
1053
|
Boué S, Talikka M, Westra JW, Hayes W, Di Fabio A, Park J, Schlage WK, Sewer A, Fields B, Ansari S, Martin F, Veljkovic E, Kenney R, Peitsch MC, Hoeng J. Causal biological network database: a comprehensive platform of causal biological network models focused on the pulmonary and vascular systems. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2015; 2015:bav030. [PMID: 25887162 PMCID: PMC4401337 DOI: 10.1093/database/bav030] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/09/2015] [Indexed: 01/28/2023]
Abstract
With the wealth of publications and data available, powerful and transparent computational approaches are required to represent measured data and scientific knowledge in a computable and searchable format. We developed a set of biological network models, scripted in the Biological Expression Language, that reflect causal signaling pathways across a wide range of biological processes, including cell fate, cell stress, cell proliferation, inflammation, tissue repair and angiogenesis in the pulmonary and cardiovascular context. This comprehensive collection of networks is now freely available to the scientific community in a centralized web-based repository, the Causal Biological Network database, which is composed of over 120 manually curated and well annotated biological network models and can be accessed at http://causalbionet.com. The website accesses a MongoDB, which stores all versions of the networks as JSON objects and allows users to search for genes, proteins, biological processes, small molecules and keywords in the network descriptions to retrieve biological networks of interest. The content of the networks can be visualized and browsed. Nodes and edges can be filtered and all supporting evidence for the edges can be browsed and is linked to the original articles in PubMed. Moreover, networks may be downloaded for further visualization and evaluation. Database URL:http://causalbionet.com
Collapse
Affiliation(s)
- Stéphanie Boué
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Marja Talikka
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Jurjen Willem Westra
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - William Hayes
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Anselmo Di Fabio
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Jennifer Park
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Walter K Schlage
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Alain Sewer
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Brett Fields
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Sam Ansari
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Florian Martin
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Emilija Veljkovic
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Renee Kenney
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Manuel C Peitsch
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| | - Julia Hoeng
- Philip Morris International R&D, Philip Morris Products S.A. Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, Selventa, One Alewife Center, Cambridge, MA 02140, USA and Applied Dynamic Solutions, LLC, 220 Davidson Avenue, Suite 100, Somerset, NJ 08873, USA
| |
Collapse
|
1054
|
Hernansaiz-Ballesteros RD, Salavert F, Sebastián-León P, Alemán A, Medina I, Dopazo J. Assessing the impact of mutations found in next generation sequencing data over human signaling pathways. Nucleic Acids Res 2015; 43:W270-5. [PMID: 25883139 PMCID: PMC4489259 DOI: 10.1093/nar/gkv349] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 04/02/2015] [Indexed: 01/20/2023] Open
Abstract
Modern sequencing technologies produce increasingly detailed data on genomic variation. However, conventional methods for relating either individual variants or mutated genes to phenotypes present known limitations given the complex, multigenic nature of many diseases or traits. Here we present PATHiVar, a web-based tool that integrates genomic variation data with gene expression tissue information. PATHiVar constitutes a new generation of genomic data analysis methods that allow studying variants found in next generation sequencing experiment in the context of signaling pathways. Simple Boolean models of pathways provide detailed descriptions of the impact of mutations in cell functionality so as, recurrences in functionality failures can easily be related to diseases, even if they are produced by mutations in different genes. Patterns of changes in signal transmission circuits, often unpredictable from individual genes mutated, correspond to patterns of affected functionalities that can be related to complex traits such as disease progression, drug response, etc. PATHiVar is available at: http://pathivar.babelomics.org.
Collapse
Affiliation(s)
| | - Francisco Salavert
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, 46012, Spain Bioinformatics of Rare Diseases (BIER), CIBER de Enfermedades Raras (CIBERER), Valencia, 46012, Spain
| | - Patricia Sebastián-León
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, 46012, Spain
| | - Alejandro Alemán
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, 46012, Spain Bioinformatics of Rare Diseases (BIER), CIBER de Enfermedades Raras (CIBERER), Valencia, 46012, Spain
| | - Ignacio Medina
- HPC Services, University of Cambridge, Cambridge, CB3 0RB, UK
| | - Joaquín Dopazo
- Computational Genomics Department, Centro de Investigación Príncipe Felipe (CIPF), Valencia, 46012, Spain Bioinformatics of Rare Diseases (BIER), CIBER de Enfermedades Raras (CIBERER), Valencia, 46012, Spain Functional Genomics Node, (INB) at CIPF, Valencia, 45012, Spain
| |
Collapse
|
1055
|
Piñero J, Queralt-Rosinach N, Bravo À, Deu-Pons J, Bauer-Mehren A, Baron M, Sanz F, Furlong LI. DisGeNET: a discovery platform for the dynamical exploration of human diseases and their genes. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2015; 2015:bav028. [PMID: 25877637 PMCID: PMC4397996 DOI: 10.1093/database/bav028] [Citation(s) in RCA: 630] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 03/09/2015] [Indexed: 11/25/2022]
Abstract
DisGeNET is a comprehensive discovery platform designed to address a variety of questions concerning the genetic underpinning of human diseases. DisGeNET contains over 380 000 associations between >16 000 genes and 13 000 diseases, which makes it one of the largest repositories currently available of its kind. DisGeNET integrates expert-curated databases with text-mined data, covers information on Mendelian and complex diseases, and includes data from animal disease models. It features a score based on the supporting evidence to prioritize gene-disease associations. It is an open access resource available through a web interface, a Cytoscape plugin and as a Semantic Web resource. The web interface supports user-friendly data exploration and navigation. DisGeNET data can also be analysed via the DisGeNET Cytoscape plugin, and enriched with the annotations of other plugins of this popular network analysis software suite. Finally, the information contained in DisGeNET can be expanded and complemented using Semantic Web technologies and linked to a variety of resources already present in the Linked Data cloud. Hence, DisGeNET offers one of the most comprehensive collections of human gene-disease associations and a valuable set of tools for investigating the molecular mechanisms underlying diseases of genetic origin, designed to fulfill the needs of different user profiles, including bioinformaticians, biologists and health-care practitioners. Database URL: http://www.disgenet.org/
Collapse
Affiliation(s)
- Janet Piñero
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain, Roche Pharma Research and Early Development, pRED Informatics, Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany and Scientific & Business Information Services, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Núria Queralt-Rosinach
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain, Roche Pharma Research and Early Development, pRED Informatics, Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany and Scientific & Business Information Services, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Àlex Bravo
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain, Roche Pharma Research and Early Development, pRED Informatics, Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany and Scientific & Business Information Services, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Jordi Deu-Pons
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain, Roche Pharma Research and Early Development, pRED Informatics, Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany and Scientific & Business Information Services, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Anna Bauer-Mehren
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain, Roche Pharma Research and Early Development, pRED Informatics, Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany and Scientific & Business Information Services, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Martin Baron
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain, Roche Pharma Research and Early Development, pRED Informatics, Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany and Scientific & Business Information Services, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Ferran Sanz
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain, Roche Pharma Research and Early Development, pRED Informatics, Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany and Scientific & Business Information Services, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Laura I Furlong
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain, Roche Pharma Research and Early Development, pRED Informatics, Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany and Scientific & Business Information Services, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| |
Collapse
|
1056
|
Durmuş S, Çakır T, Özgür A, Guthke R. A review on computational systems biology of pathogen-host interactions. Front Microbiol 2015; 6:235. [PMID: 25914674 PMCID: PMC4391036 DOI: 10.3389/fmicb.2015.00235] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/10/2015] [Indexed: 12/27/2022] Open
Abstract
Pathogens manipulate the cellular mechanisms of host organisms via pathogen-host interactions (PHIs) in order to take advantage of the capabilities of host cells, leading to infections. The crucial role of these interspecies molecular interactions in initiating and sustaining infections necessitates a thorough understanding of the corresponding mechanisms. Unlike the traditional approach of considering the host or pathogen separately, a systems-level approach, considering the PHI system as a whole is indispensable to elucidate the mechanisms of infection. Following the technological advances in the post-genomic era, PHI data have been produced in large-scale within the last decade. Systems biology-based methods for the inference and analysis of PHI regulatory, metabolic, and protein-protein networks to shed light on infection mechanisms are gaining increasing demand thanks to the availability of omics data. The knowledge derived from the PHIs may largely contribute to the identification of new and more efficient therapeutics to prevent or cure infections. There are recent efforts for the detailed documentation of these experimentally verified PHI data through Web-based databases. Despite these advances in data archiving, there are still large amounts of PHI data in the biomedical literature yet to be discovered, and novel text mining methods are in development to unearth such hidden data. Here, we review a collection of recent studies on computational systems biology of PHIs with a special focus on the methods for the inference and analysis of PHI networks, covering also the Web-based databases and text-mining efforts to unravel the data hidden in the literature.
Collapse
Affiliation(s)
- Saliha Durmuş
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University, KocaeliTurkey
| | - Tunahan Çakır
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University, KocaeliTurkey
| | - Arzucan Özgür
- Department of Computer Engineering, Boǧaziçi University, IstanbulTurkey
| | - Reinhard Guthke
- Leibniz Institute for Natural Product Research and Infection Biology – Hans-Knoell-Institute, JenaGermany
| |
Collapse
|
1057
|
Campbell CL, Torres-Perez F, Acuna-Retamar M, Schountz T. Transcriptome markers of viral persistence in naturally-infected andes virus (bunyaviridae) seropositive long-tailed pygmy rice rats. PLoS One 2015; 10:e0122935. [PMID: 25856432 PMCID: PMC4391749 DOI: 10.1371/journal.pone.0122935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/24/2015] [Indexed: 12/22/2022] Open
Abstract
Long-tailed pygmy rice rats (Oligoryzomys longicaudatus) are principal reservoir hosts of Andes virus (ANDV) (Bunyaviridae), which causes most hantavirus cardiopulmonary syndrome cases in the Americas. To develop tools for the study of the ANDV-host interactions, we used RNA-Seq to generate a de novo transcriptome assembly. Splenic RNA from five rice rats captured in Chile, three of which were ANDV-infected, was used to generate an assembly of 66,173 annotated transcripts, including noncoding RNAs. Phylogenetic analysis of selected predicted proteins showed similarities to those of the North American deer mouse (Peromyscus maniculatus), the principal reservoir of Sin Nombre virus (SNV). One of the infected rice rats had about 50-fold more viral burden than the others, suggesting acute infection, whereas the remaining two had levels consistent with persistence. Differential expression analysis revealed distinct signatures among the infected rodents. The differences could be due to 1) variations in viral load, 2) dimorphic or reproductive differences in splenic homing of immune cells, or 3) factors of unknown etiology. In the two persistently infected rice rats, suppression of the JAK-STAT pathway at Stat5b and Ccnot1, elevation of Casp1, RIG-I pathway factors Ppp1cc and Mff, and increased FC receptor-like transcripts occurred. Caspase-1 and Stat5b activation pathways have been shown to stimulate T helper follicular cell (TFH) development in other species. These data are also consistent with reports suggestive of TFH stimulation in deer mice experimentally infected with hantaviruses. In the remaining acutely infected rice rat, the apoptotic pathway marker Cox6a1 was elevated, and putative anti-viral factors Abcb1a, Fam46c, Spp1, Rxra, Rxrb, Trmp2 and Trim58 were modulated. Transcripts for preproenkephalin (Prenk) were reduced, which may be predictive of an increased T cell activation threshold. Taken together, this transcriptome dataset will permit rigorous examination of rice rat-ANDV interactions and may lead to better understanding of virus ecology.
Collapse
Affiliation(s)
- Corey L. Campbell
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| | - Fernando Torres-Perez
- Instituto de Biología, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | | | - Tony Schountz
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
1058
|
Squizzato S, Park YM, Buso N, Gur T, Cowley A, Li W, Uludag M, Pundir S, Cham JA, McWilliam H, Lopez R. The EBI Search engine: providing search and retrieval functionality for biological data from EMBL-EBI. Nucleic Acids Res 2015; 43:W585-8. [PMID: 25855807 PMCID: PMC4489232 DOI: 10.1093/nar/gkv316] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/28/2015] [Indexed: 01/20/2023] Open
Abstract
The European Bioinformatics Institute (EMBL-EBI-https://www.ebi.ac.uk) provides free and unrestricted access to data across all major areas of biology and biomedicine. Searching and extracting knowledge across these domains requires a fast and scalable solution that addresses the requirements of domain experts as well as casual users. We present the EBI Search engine, referred to here as 'EBI Search', an easy-to-use fast text search and indexing system with powerful data navigation and retrieval capabilities. API integration provides access to analytical tools, allowing users to further investigate the results of their search. The interconnectivity that exists between data resources at EMBL-EBI provides easy, quick and precise navigation and a better understanding of the relationship between different data types including sequences, genes, gene products, proteins, protein domains, protein families, enzymes and macromolecular structures, together with relevant life science literature.
Collapse
Affiliation(s)
- Silvano Squizzato
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Young Mi Park
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Nicola Buso
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Tamer Gur
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Andrew Cowley
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Weizhong Li
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Mahmut Uludag
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Sangya Pundir
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Jennifer A Cham
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Hamish McWilliam
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| | - Rodrigo Lopez
- European Bioinformatics Institute, EMBL Outstation, Wellcome Trust Genome Campus, Hinxton, CB10 1SD, Cambridge, UK
| |
Collapse
|
1059
|
Paiva C, Amaral A, Rodriguez M, Canyellas N, Correig X, Ballescà JL, Ramalho-Santos J, Oliva R. Identification of endogenous metabolites in human sperm cells using proton nuclear magnetic resonance ((1) H-NMR) spectroscopy and gas chromatography-mass spectrometry (GC-MS). Andrology 2015; 3:496-505. [PMID: 25854681 DOI: 10.1111/andr.12027] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/26/2015] [Accepted: 02/09/2015] [Indexed: 12/15/2022]
Abstract
The objective of this study was to contribute to the first comprehensive metabolomic characterization of the human sperm cell through the application of two untargeted platforms based on proton nuclear magnetic resonance ((1) H-NMR) spectroscopy and gas chromatography coupled to mass spectrometry (GC-MS). Using these two complementary strategies, we were able to identify a total of 69 metabolites, of which 42 were identified using NMR, 27 using GC-MS and 4 by both techniques. The identity of some of these metabolites was further confirmed by two-dimensional (1) H-(1) H homonuclear correlation spectroscopy (COSY) and (1) H-(13) C heteronuclear single-quantum correlation (HSQC) spectroscopy. Most of the metabolites identified are reported here for the first time in mature human spermatozoa. The relationship between the metabolites identified and the previously reported sperm proteome was also explored. Interestingly, overrepresented pathways included not only the metabolism of carbohydrates, but also of lipids and lipoproteins. Of note, a large number of the metabolites identified belonged to the amino acids, peptides and analogues super class. The identification of this initial set of metabolites represents an important first step to further study their function in male gamete physiology and to explore potential reasons for dysfunction in future studies. We also demonstrate that the application of NMR and MS provides complementary results, thus constituting a promising strategy towards the completion of the human sperm cell metabolome.
Collapse
Affiliation(s)
- C Paiva
- Faculty of Medicine, Human Genetics Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biochemistry and Molecular Genetics Service, Hospital Clinic, Barcelona, Spain.,Biology of Reproduction and Stem Cell Group, CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,PhD Program in Experimental Biology and Biomedicine (PDBEB), Center for Neuroscience and Cell Biology, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - A Amaral
- Faculty of Medicine, Human Genetics Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biochemistry and Molecular Genetics Service, Hospital Clinic, Barcelona, Spain.,Biology of Reproduction and Stem Cell Group, CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - M Rodriguez
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Institut d'Investigació Sanitària Pere Virgili (IISPV) and Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - N Canyellas
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Institut d'Investigació Sanitària Pere Virgili (IISPV) and Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - X Correig
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Institut d'Investigació Sanitària Pere Virgili (IISPV) and Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - J L Ballescà
- Clinic Institute of Gynaecology, Obstetrics and Neonatology, Hospital Clinic, Barcelona, Spain
| | - J Ramalho-Santos
- Biology of Reproduction and Stem Cell Group, CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - R Oliva
- Faculty of Medicine, Human Genetics Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biochemistry and Molecular Genetics Service, Hospital Clinic, Barcelona, Spain
| |
Collapse
|
1060
|
Scott CC, Vossio S, Vacca F, Snijder B, Larios J, Schaad O, Guex N, Kuznetsov D, Martin O, Chambon M, Turcatti G, Pelkmans L, Gruenberg J. Wnt directs the endosomal flux of LDL-derived cholesterol and lipid droplet homeostasis. EMBO Rep 2015; 16:741-52. [PMID: 25851648 DOI: 10.15252/embr.201540081] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/06/2015] [Indexed: 01/24/2023] Open
Abstract
The Wnt pathway, which controls crucial steps of the development and differentiation programs, has been proposed to influence lipid storage and homeostasis. In this paper, using an unbiased strategy based on high-content genome-wide RNAi screens that monitored lipid distribution and amounts, we find that Wnt3a regulates cellular cholesterol. We show that Wnt3a stimulates the production of lipid droplets and that this stimulation strictly depends on endocytosed, LDL-derived cholesterol and on functional early and late endosomes. We also show that Wnt signaling itself controls cholesterol endocytosis and flux along the endosomal pathway, which in turn modulates cellular lipid homeostasis. These results underscore the importance of endosome functions for LD formation and reveal a previously unknown regulatory mechanism of the cellular programs controlling lipid storage and endosome transport under the control of Wnt signaling.
Collapse
Affiliation(s)
- Cameron C Scott
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Stefania Vossio
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Fabrizio Vacca
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Berend Snijder
- Faculty of Sciences, Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Jorge Larios
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Olivier Schaad
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Nicolas Guex
- Vital-IT Group, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - Dmitry Kuznetsov
- Vital-IT Group, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - Olivier Martin
- Vital-IT Group, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - Marc Chambon
- Biomolecular Screening Facility, SV-PTECH-PTCB, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Gerardo Turcatti
- Biomolecular Screening Facility, SV-PTECH-PTCB, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Lucas Pelkmans
- Faculty of Sciences, Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Jean Gruenberg
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| |
Collapse
|
1061
|
Gu L, Evans AR, Robinson RAS. Sample multiplexing with cysteine-selective approaches: cysDML and cPILOT. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:615-630. [PMID: 25588721 DOI: 10.1007/s13361-014-1059-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 11/22/2014] [Accepted: 11/22/2014] [Indexed: 06/04/2023]
Abstract
Cysteine-selective proteomics approaches simplify complex protein mixtures and improve the chance of detecting low abundant proteins. It is possible that cysteinyl-peptide/protein enrichment methods could be coupled to isotopic labeling and isobaric tagging methods for quantitative proteomics analyses in as few as two or up to 10 samples, respectively. Here we present two novel cysteine-selective proteomics approaches: cysteine-selective dimethyl labeling (cysDML) and cysteine-selective combined precursor isotopic labeling and isobaric tagging (cPILOT). CysDML is a duplex precursor quantification technique that couples cysteinyl-peptide enrichment with on-resin stable-isotope dimethyl labeling. Cysteine-selective cPILOT is a novel 12-plex workflow based on cysteinyl-peptide enrichment, on-resin stable-isotope dimethyl labeling, and iodoTMT tagging on cysteine residues. To demonstrate the broad applicability of the approaches, we applied cysDML and cPILOT methods to liver tissues from an Alzheimer's disease (AD) mouse model and wild-type (WT) controls. From the cysDML experiments, an average of 850 proteins were identified and 594 were quantified, whereas from the cPILOT experiment, 330 and 151 proteins were identified and quantified, respectively. Overall, 2259 unique total proteins were detected from both cysDML and cPILOT experiments. There is tremendous overlap in the proteins identified and quantified between both experiments, and many proteins have AD/WT fold-change values that are within ~20% error. A total of 65 statistically significant proteins are differentially expressed in the liver proteome of AD mice relative to WT. The performance of cysDML and cPILOT are demonstrated and advantages and limitations of using multiple duplex experiments versus a single 12-plex experiment are highlighted.
Collapse
Affiliation(s)
- Liqing Gu
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | | | | |
Collapse
|
1062
|
Abstract
An important data analysis task in statistical genomics involves the integration of genome-wide gene-level measurements with preexisting data on the same genes. A wide variety of statistical methodologies and computational tools have been developed for this general task. We emphasize one particular distinction among methodologies, namely whether they process gene sets one at a time (uniset) or simultaneously via some multiset technique. Owing to the complexity of collections of gene sets, the multiset approach offers some advantages, as it naturally accommodates set-size variations and among-set overlaps. However, this approach presents both computational and inferential challenges. After reviewing some statistical issues that arise in uniset analysis, we examine two model-based multiset methods for gene list data.
Collapse
Affiliation(s)
- Michael A Newton
- Department of Statistics, University of Wisconsin, Madison, Wisconsin 53706 ; Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin 53706
| | - Zhishi Wang
- Department of Statistics, University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
1063
|
Abstract
Accurate identification of drug targets is a crucial part of any drug development program. We mined the human proteome to discover properties of proteins that may be important in determining their suitability for pharmaceutical modulation. Data was gathered concerning each protein's sequence, post-translational modifications, secondary structure, germline variants, expression profile and drug target status. The data was then analysed to determine features for which the target and non-target proteins had significantly different values. This analysis was repeated for subsets of the proteome consisting of all G-protein coupled receptors, ion channels, kinases and proteases, as well as proteins that are implicated in cancer. Machine learning was used to quantify the proteins in each dataset in terms of their potential to serve as a drug target. This was accomplished by first inducing a random forest that could distinguish between its targets and non-targets, and then using the random forest to quantify the drug target likeness of the non-targets. The properties that can best differentiate targets from non-targets were primarily those that are directly related to a protein's sequence (e.g. secondary structure). Germline variants, expression levels and interactions between proteins had minimal discriminative power. Overall, the best indicators of drug target likeness were found to be the proteins' hydrophobicities, in vivo half-lives, propensity for being membrane bound and the fraction of non-polar amino acids in their sequences. In terms of predicting potential targets, datasets of proteases, ion channels and cancer proteins were able to induce random forests that were highly capable of distinguishing between targets and non-targets. The non-target proteins predicted to be targets by these random forests comprise the set of the most suitable potential future drug targets, and should therefore be prioritised when building a drug development programme.
Collapse
Affiliation(s)
- Simon C. Bull
- Manchester Institute of Biotechnology, Faculty of Life Sciences, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kigndom
| | - Andrew J. Doig
- Manchester Institute of Biotechnology, Faculty of Life Sciences, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kigndom
| |
Collapse
|
1064
|
Increased expression of interferon signaling genes in the bone marrow microenvironment of myelodysplastic syndromes. PLoS One 2015; 10:e0120602. [PMID: 25803272 PMCID: PMC4372597 DOI: 10.1371/journal.pone.0120602] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 01/24/2015] [Indexed: 11/19/2022] Open
Abstract
Introduction The bone marrow (BM) microenvironment plays an important role in the pathogenesis of myelodysplastic syndromes (MDS) through a reciprocal interaction with resident BM hematopoietic cells. We investigated the differences between BM mesenchymal stromal cells (MSCs) in MDS and normal individuals and identified genes involved in such differences. Materials and Methods BM-derived MSCs from 7 MDS patients (3 RCMD, 3 RAEB-1, and 1 RAEB-2) and 7 controls were cultured. Global gene expression was analyzed using a microarray. Result We found 314 differentially expressed genes (DEGs) in RCMD vs. control, 68 in RAEB vs. control, and 51 in RAEB vs. RCMD. All comparisons were clearly separated from one another by hierarchical clustering. The overall similarity between differential expression signatures from the RCMD vs. control comparison and the RAEB vs. control comparison was highly significant (p = 0), which indicates a common transcriptomic response in these two MDS subtypes. RCMD and RAEB simultaneously showed an up-regulation of interferon alpha/beta signaling and the ISG15 antiviral mechanism, and a significant fraction of the RAEB vs. control DEGs were also putative targets of transcription factors IRF and ICSBP. Pathways that involved RNA polymerases I and III and mitochondrial transcription were down-regulated in RAEB compared to RCMD. Conclusion Gene expression in the MDS BM microenvironment was different from that in normal BM and exhibited altered expression according to disease progression. The present study provides genetic evidence that inflammation and immune dysregulation responses that involve the interferon signaling pathway in the BM microenvironment are associated with MDS pathogenesis, which suggests BM MSCs as a possible therapeutic target in MDS.
Collapse
|
1065
|
Polotskaia A, Xiao G, Reynoso K, Martin C, Qiu WG, Hendrickson RC, Bargonetti J. Proteome-wide analysis of mutant p53 targets in breast cancer identifies new levels of gain-of-function that influence PARP, PCNA, and MCM4. Proc Natl Acad Sci U S A 2015; 112:E1220-9. [PMID: 25733866 PMCID: PMC4371979 DOI: 10.1073/pnas.1416318112] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The gain-of-function mutant p53 (mtp53) transcriptome has been studied, but, to date, no detailed analysis of the mtp53-associated proteome has been described. We coupled cell fractionation with stable isotope labeling with amino acids in cell culture (SILAC) and inducible knockdown of endogenous mtp53 to determine the mtp53-driven proteome. Our fractionation data highlight the underappreciated biology that missense mtp53 proteins R273H, R280K, and L194F are tightly associated with chromatin. Using SILAC coupled to tandem MS, we identified that R273H mtp53 expression in MDA-MB-468 breast cancer cells up- and down-regulated multiple proteins and metabolic pathways. Here we provide the data set obtained from sequencing 73,154 peptide pairs that then corresponded to 3,010 proteins detected under reciprocal labeling conditions. Importantly, the high impact regulated targets included the previously identified transcriptionally regulated mevalonate pathway proteins but also identified two new levels of mtp53 protein regulation for nontranscriptional targets. Interestingly, mtp53 depletion profoundly influenced poly(ADP ribose) polymerase 1 (PARP1) localization, with increased cytoplasmic and decreased chromatin-associated protein. An enzymatic PARP shift occurred with high mtp53 expression, resulting in increased poly-ADP-ribosylated proteins in the nucleus. Mtp53 increased the level of proliferating cell nuclear antigen (PCNA) and minichromosome maintenance 4 (MCM4) proteins without changing the amount of pcna and mcm4 transcripts. Pathway enrichment analysis ranked the DNA replication pathway above the cholesterol biosynthesis pathway as a R273H mtp53 activated proteomic target. Knowledge of the proteome diversity driven by mtp53 suggests that DNA replication and repair pathways are major targets of mtp53 and highlights consideration of combination chemotherapeutic strategies targeting cholesterol biosynthesis and PARP inhibition.
Collapse
Affiliation(s)
- Alla Polotskaia
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Gu Xiao
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Katherine Reynoso
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Che Martin
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Wei-Gang Qiu
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Ronald C Hendrickson
- Proteomics Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Jill Bargonetti
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| |
Collapse
|
1066
|
Rajput NK, Singh V, Bhardwaj A. Resources, challenges and way forward in rare mitochondrial diseases research. F1000Res 2015; 4:70. [PMID: 26180633 DOI: 10.12688/f1000research.6208.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/13/2015] [Indexed: 12/27/2022] Open
Abstract
Over 300 million people are affected by about 7000 rare diseases globally. There are tremendous resource limitations and challenges in driving research and drug development for rare diseases. Hence, innovative approaches are needed to identify potential solutions. This review focuses on the resources developed over the past years for analysis of genome data towards understanding disease biology especially in the context of mitochondrial diseases, given that mitochondria are central to major cellular pathways and their dysfunction leads to a broad spectrum of diseases. Platforms for collaboration of research groups, clinicians and patients and the advantages of community collaborative efforts in addressing rare diseases are also discussed. The review also describes crowdsourcing and crowdfunding efforts in rare diseases research and how the upcoming initiatives for understanding disease biology including analyses of large number of genomes are also applicable to rare diseases.
Collapse
Affiliation(s)
- Neeraj Kumar Rajput
- Open Source Drug Discovery (OSDD) Unit, Council of Scientific and Industrial Research, New Delhi, 110001, India
| | - Vipin Singh
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, 201301, India
| | - Anshu Bhardwaj
- Open Source Drug Discovery (OSDD) Unit, Council of Scientific and Industrial Research, New Delhi, 110001, India
| |
Collapse
|
1067
|
Li J, Chen H, Ren J, Song J, Zhang F, Zhang J, Lee C, Li S, Geng Q, Cao C, Xu N. Effects of statin on circulating microRNAome and predicted function regulatory network in patients with unstable angina. BMC Med Genomics 2015; 8:12. [PMID: 25889164 PMCID: PMC4364658 DOI: 10.1186/s12920-015-0082-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/06/2015] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Statin therapy plays a pivotal role in stabilizing the plaque for unstable angina (UA) patients although its mechanism(s) remains largely unexplored. Here we aim to identify microRNAs (miRNAs) mediating the protective effect of statins in UA patients. METHODS MiRNAs Array was carried out to compare the circulating whole blood miRNA profile of UA patients treated with (n = 10) and without statin (n = 10) and plasma miRNA profile UA patients treated with (n = 5) and without statin (n = 5). 22 whole blood miRNAs and 19 plasma miRNAs were found significantly upregulated in statin group. Targets of these miRNAs were predicted by algoritms: Targetscan, Miranda and Diana microT, then clustered according to functions and cell types by using the Database for Annotation, Visualization and Integrated Discovery (DAVID). To reveal the enriched function pathways in human atherosclerotic plaque, we analyzed microarray data from GEO database, Coronary atherosclerotic plaque (n = 80); macrophages in ruptured plaque (n = 11); carotid atheroma plaque (n = 64); advanced carotid atherosclerotic plaque (n = 29) using Reactome database. Integrated analysis indicated that statin induced miRNAs mainly regulate the signaling pathways of Rho GTPase and hemostasis in human atherosclerotic lesion. In vulnerable plaque, additional immune system signaling was also targeted. RESULTS The data showed target genes regulated by these statin induced miRNAs majorly expressed in i) plaque macrophage and platelet, where they were involved in hemostasis process; ii) in monocyte to regulate NGF apoptosis; iii) and in endothelial cell function in Rho GTPase pathway. Integrate analysis indicated that statin induced miRNAs mainly regulate the signaling pathways of Rho GTPase and hemostasis in human atherosclerotic lesion. CONCLUSIONS Our study suggest that statin induces the expression of multiple miRNAs in the circulation of UA patient, which play important roles by regulating signal pathways critical for the pathogenesis of UA.
Collapse
Affiliation(s)
- Jingjin Li
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Hong Chen
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Jingyi Ren
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Junxian Song
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Feng Zhang
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Jing Zhang
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Chongyou Lee
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Sufang Li
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Qiang Geng
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Chengfu Cao
- Department of Cardiology, Peking University People's hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
| | - Ning Xu
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
1068
|
Nishioka T, Shohag MH, Amano M, Kaibuchi K. Developing novel methods to search for substrates of protein kinases such as Rho-kinase. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1663-6. [PMID: 25770685 DOI: 10.1016/j.bbapap.2015.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/05/2015] [Indexed: 01/18/2023]
Abstract
Protein phosphorylation is a major and essential post-translational modification in eukaryotic cells that plays a critical role in various cellular processes. Recent progresses in mass spectrometry techniques have enabled the effective identification and analysis of protein phosphorylation. Mass spectrometry-based approaches in investigating protein phosphorylation are very powerful and informative and can further improve our understanding of protein phosphorylation as a whole, but they cannot determine the upstream kinases involved. We introduce several studies that attempted to uncover the relationships between various kinases of interest and substrates, including two methods we developed: an in vitro approach termed the kinase-interacting substrate screening (KISS) method and an in vivo approach termed the phosphatase inhibitor and kinase inhibitor substrate screening (PIKISS) method. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.
Collapse
Affiliation(s)
- Tomoki Nishioka
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Md Hasanuzzaman Shohag
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Mutsuki Amano
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan.
| |
Collapse
|
1069
|
Li J, Chanrion M, Sawey E, Wang T, Chow E, Tward A, Su Y, Xue W, Lucito R, Zender L, Lowe SW, Bishop JM, Powers S. Reciprocal interaction of Wnt and RXR-α pathways in hepatocyte development and hepatocellular carcinoma. PLoS One 2015; 10:e0118480. [PMID: 25738607 PMCID: PMC4349704 DOI: 10.1371/journal.pone.0118480] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 01/14/2015] [Indexed: 11/29/2022] Open
Abstract
Genomic analysis of human hepatocellular carcinoma (HCC) is potentially confounded by the differentiation state of the hepatic cell-of-origin. Here we integrated genomic analysis of mouse HCC (with defined cell-of-origin) along with normal development. We found a major shift in expression of Wnt and RXR-α pathway genes (up and down, respectively) coincident with the transition from hepatoblasts to hepatocytes. A combined Wnt and RXR-α gene signature categorized HCCs into two subtypes (high Wnt, low RXR-α and low Wnt, high RXR-α), which matched cell-of-origin in mouse models and the differentiation state of human HCC. Suppression of RXR-α levels in hepatocytes increased Wnt signaling and enhanced tumorigenicity, whereas ligand activation of RXR-α achieved the opposite. These results corroborate that there are two main HCC subtypes that correspond to the degree of hepatocyte differentation and that RXR-α, in part via Wnt signaling, plays a key functional role in the hepatocyte-like subtype and potentially could serve as a selective therapeutic target.
Collapse
Affiliation(s)
- Jinyu Li
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Maia Chanrion
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Eric Sawey
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Tim Wang
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Edward Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Aaron Tward
- G. W. Hooper Foundation and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, United States of America
| | - Yi Su
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Wen Xue
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, United States of America
| | - Robert Lucito
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
| | - Lars Zender
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, United States of America
| | - Scott W. Lowe
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, United States of America
| | - J. Michael Bishop
- G. W. Hooper Foundation and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, United States of America
| | - Scott Powers
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY 11740, United States of America
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, United States of America
- * E-mail:
| |
Collapse
|
1070
|
Abstract
Behaviours of complex biomolecular systems are often irreducible to the elementary properties of their individual components. Explanatory and predictive mathematical models are therefore useful for fully understanding and precisely engineering cellular functions. The development and analyses of these models require their adaptation to the problems that need to be solved and the type and amount of available genetic or molecular data. Quantitative and logic modelling are among the main methods currently used to model molecular and gene networks. Each approach comes with inherent advantages and weaknesses. Recent developments show that hybrid approaches will become essential for further progress in synthetic biology and in the development of virtual organisms.
Collapse
Affiliation(s)
- Nicolas Le Novère
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| |
Collapse
|
1071
|
Zou D, Ma L, Yu J, Zhang Z. Biological databases for human research. GENOMICS PROTEOMICS & BIOINFORMATICS 2015; 13:55-63. [PMID: 25712261 PMCID: PMC4411498 DOI: 10.1016/j.gpb.2015.01.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 01/01/2023]
Abstract
The completion of the Human Genome Project lays a foundation for systematically studying the human genome from evolutionary history to precision medicine against diseases. With the explosive growth of biological data, there is an increasing number of biological databases that have been developed in aid of human-related research. Here we present a collection of human-related biological databases and provide a mini-review by classifying them into different categories according to their data types. As human-related databases continue to grow not only in count but also in volume, challenges are ahead in big data storage, processing, exchange and curation.
Collapse
Affiliation(s)
- Dong Zou
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lina Ma
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Yu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zhang Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
1072
|
Mills CL, Beuning PJ, Ondrechen MJ. Biochemical functional predictions for protein structures of unknown or uncertain function. Comput Struct Biotechnol J 2015; 13:182-91. [PMID: 25848497 PMCID: PMC4372640 DOI: 10.1016/j.csbj.2015.02.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 02/06/2015] [Accepted: 02/11/2015] [Indexed: 01/07/2023] Open
Abstract
With the exponential growth in the determination of protein sequences and structures via genome sequencing and structural genomics efforts, there is a growing need for reliable computational methods to determine the biochemical function of these proteins. This paper reviews the efforts to address the challenge of annotating the function at the molecular level of uncharacterized proteins. While sequence- and three-dimensional-structure-based methods for protein function prediction have been reviewed previously, the recent trends in local structure-based methods have received less attention. These local structure-based methods are the primary focus of this review. Computational methods have been developed to predict the residues important for catalysis and the local spatial arrangements of these residues can be used to identify protein function. In addition, the combination of different types of methods can help obtain more information and better predictions of function for proteins of unknown function. Global initiatives, including the Enzyme Function Initiative (EFI), COMputational BRidges to EXperiments (COMBREX), and the Critical Assessment of Function Annotation (CAFA), are evaluating and testing the different approaches to predicting the function of proteins of unknown function. These initiatives and global collaborations will increase the capability and reliability of methods to predict biochemical function computationally and will add substantial value to the current volume of structural genomics data by reducing the number of absent or inaccurate functional annotations.
Collapse
Affiliation(s)
- Caitlyn L Mills
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, United States
| | - Penny J Beuning
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, United States
| | - Mary Jo Ondrechen
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, United States
| |
Collapse
|
1073
|
Kuperstein I, Grieco L, Cohen DPA, Thieffry D, Zinovyev A, Barillot E. The shortest path is not the one you know: application of biological network resources in precision oncology research. Mutagenesis 2015; 30:191-204. [DOI: 10.1093/mutage/geu078] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
1074
|
Galligan J, Martinez-Noël G, Arndt V, Hayes S, Chittenden TW, Harper JW, Howley PM. Proteomic analysis and identification of cellular interactors of the giant ubiquitin ligase HERC2. J Proteome Res 2015; 14:953-66. [PMID: 25476789 PMCID: PMC4324439 DOI: 10.1021/pr501005v] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Indexed: 01/10/2023]
Abstract
HERC2 is a large E3 ubiquitin ligase with multiple structural domains that has been implicated in an array of cellular processes. Mutations in HERC2 are linked to developmental delays and impairment caused by nervous system dysfunction, such as Angelman Syndrome and autism-spectrum disorders. However, HERC2 cellular activity and regulation remain poorly understood. We used a broad proteomic approach to survey the landscape of cellular proteins that interact with HERC2. We identified nearly 300 potential interactors, a subset of which we validated binding to HERC2. The potential HERC2 interactors included the eukaryotic translation initiation factor 3 complex, the intracellular transport COPI coatomer complex, the glycogen regulator phosphorylase kinase, beta-catenin, PI3 kinase, and proteins involved in fatty acid transport and iron homeostasis. Through a complex bioinformatic analysis of potential interactors, we linked HERC2 to cellular processes including intracellular protein trafficking and transport, metabolism of cellular energy, and protein translation. Given its size, multidomain structure, and association with various cellular activities, HERC2 may function as a scaffold to integrate protein complexes and bridge critical cellular pathways. This work provides a significant resource with which to interrogate HERC2 function more deeply and evaluate its contributions to mechanisms governing cellular homeostasis and disease.
Collapse
Affiliation(s)
- Jeffrey
T. Galligan
- Department
of Microbiology and Immunobiology, Harvard
Medical School, 77 Avenue
Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Gustavo Martinez-Noël
- Department
of Microbiology and Immunobiology, Harvard
Medical School, 77 Avenue
Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Verena Arndt
- Department
of Microbiology and Immunobiology, Harvard
Medical School, 77 Avenue
Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Sebastian Hayes
- Department
of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Thomas W. Chittenden
- Research
Computing Group, Harvard Medical School, 25 Shattuck Street #500, Boston, Massachusetts 02115, United States
- Complex Biological
Systems Alliance, 17 Peterson Road, North Andover, Massachusetts 01845, United States
| | - J. Wade Harper
- Department
of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Peter M. Howley
- Department
of Microbiology and Immunobiology, Harvard
Medical School, 77 Avenue
Louis Pasteur, Boston, Massachusetts 02115, United States
| |
Collapse
|
1075
|
Villaveces JM, Jiménez RC, Porras P, Del-Toro N, Duesbury M, Dumousseau M, Orchard S, Choi H, Ping P, Zong NC, Askenazi M, Habermann BH, Hermjakob H. Merging and scoring molecular interactions utilising existing community standards: tools, use-cases and a case study. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2015; 2015:bau131. [PMID: 25652942 PMCID: PMC4316181 DOI: 10.1093/database/bau131] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The evidence that two molecules interact in a living cell is often inferred from multiple different experiments. Experimental data is captured in multiple repositories, but there is no simple way to assess the evidence of an interaction occurring in a cellular environment. Merging and scoring of data are commonly required operations after querying for the details of specific molecular interactions, to remove redundancy and assess the strength of accompanying experimental evidence. We have developed both a merging algorithm and a scoring system for molecular interactions based on the proteomics standard initiative–molecular interaction standards. In this manuscript, we introduce these two algorithms and provide community access to the tool suite, describe examples of how these tools are useful to selectively present molecular interaction data and demonstrate a case where the algorithms were successfully used to identify a systematic error in an existing dataset.
Collapse
Affiliation(s)
- J M Villaveces
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - R C Jiménez
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - P Porras
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - N Del-Toro
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - M Duesbury
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - M Dumousseau
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - S Orchard
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - H Choi
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - P Ping
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - N C Zong
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - M Askenazi
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - B H Habermann
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| | - Henning Hermjakob
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Matinsried, Germany, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK, Department of Physiology and Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive, MRL Building, Suite 1609, Los Angeles, California 90095, USA and Biomedical Hosting LLC, Arlington, Massachusetts 02474, USA
| |
Collapse
|
1076
|
Forés-Martos J, Cervera-Vidal R, Chirivella E, Ramos-Jarero A, Climent J. A genomic approach to study down syndrome and cancer inverse comorbidity: untangling the chromosome 21. Front Physiol 2015; 6:10. [PMID: 25698970 PMCID: PMC4316712 DOI: 10.3389/fphys.2015.00010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 01/08/2015] [Indexed: 12/19/2022] Open
Abstract
Down syndrome (DS), one of the most common birth defects and the most widespread genetic cause of intellectual disabilities, is caused by extra genetic material on chromosome 21 (HSA21). The increased genomic dosage of trisomy 21 is thought to be responsible for the distinct DS phenotypes, including an increased risk of developing some types of childhood leukemia and germ cell tumors. Patients with DS, however, have a strikingly lower incidence of many other solid tumors. We hypothesized that the third copy of genes located in HSA21 may have an important role on the protective effect that DS patients show against most types of solid tumors. Focusing on Copy Number Variation (CNV) array data, we have generated frequencies of deleted regions in HSA21 in four different tumor types from which DS patients have been reported to be protected. We describe three different regions of deletion pointing to a set of candidate genes that could explain the inverse comorbidity phenomenon between DS and solid tumors. In particular we found RCAN1 gene in Wilms tumors and a miRNA cluster containing miR-99A, miR-125B2 and miR-LET7C in lung, breast, and melanoma tumors as the main candidates for explaining the inverse comorbidity observed between solid tumors and DS.
Collapse
Affiliation(s)
- Jaume Forés-Martos
- Genomics and Systems Biology (InGSB) Lab, Oncology and Hematology Department, Biomedical Research Institute INCLIVA Valencia, Spain
| | - Raimundo Cervera-Vidal
- Genomics and Systems Biology (InGSB) Lab, Oncology and Hematology Department, Biomedical Research Institute INCLIVA Valencia, Spain
| | - Enrique Chirivella
- Genomics and Systems Biology (InGSB) Lab, Oncology and Hematology Department, Biomedical Research Institute INCLIVA Valencia, Spain
| | - Alberto Ramos-Jarero
- Genomics and Systems Biology (InGSB) Lab, Oncology and Hematology Department, Biomedical Research Institute INCLIVA Valencia, Spain
| | - Joan Climent
- Genomics and Systems Biology (InGSB) Lab, Oncology and Hematology Department, Biomedical Research Institute INCLIVA Valencia, Spain
| |
Collapse
|
1077
|
Tsoi LC, Iyer MK, Stuart PE, Swindell WR, Gudjonsson JE, Tejasvi T, Sarkar MK, Li B, Ding J, Voorhees JJ, Kang HM, Nair RP, Chinnaiyan AM, Abecasis GR, Elder JT. Analysis of long non-coding RNAs highlights tissue-specific expression patterns and epigenetic profiles in normal and psoriatic skin. Genome Biol 2015; 16:24. [PMID: 25723451 PMCID: PMC4311508 DOI: 10.1186/s13059-014-0570-4] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/11/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Although analysis pipelines have been developed to use RNA-seq to identify long non-coding RNAs (lncRNAs), inference of their biological and pathological relevance remains a challenge. As a result, most transcriptome studies of autoimmune disease have only assessed protein-coding transcripts. RESULTS We used RNA-seq data from 99 lesional psoriatic, 27 uninvolved psoriatic, and 90 normal skin biopsies, and applied computational approaches to identify and characterize expressed lncRNAs. We detect 2,942 previously annotated and 1,080 novel lncRNAs which are expected to be skin specific. Notably, over 40% of the novel lncRNAs are differentially expressed and the proportions of differentially expressed transcripts among protein-coding mRNAs and previously-annotated lncRNAs are lower in psoriasis lesions versus uninvolved or normal skin. We find that many lncRNAs, in particular those that are differentially expressed, are co-expressed with genes involved in immune related functions, and that novel lncRNAs are enriched for localization in the epidermal differentiation complex. We also identify distinct tissue-specific expression patterns and epigenetic profiles for novel lncRNAs, some of which are shown to be regulated by cytokine treatment in cultured human keratinocytes. CONCLUSIONS Together, our results implicate many lncRNAs in the immunopathogenesis of psoriasis, and our results provide a resource for lncRNA studies in other autoimmune diseases.
Collapse
Affiliation(s)
- Lam C Tsoi
- />Department of Biostatistics, Center for Statistical Genetics, School of Public Health, M4614 SPH I, University of Michigan, Box 2029, Ann Arbor, MI 48109-2029 USA
| | - Matthew K Iyer
- />Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI USA
| | - Philip E Stuart
- />Department of Dermatology, University of Michigan, Ann Arbor, MI USA
| | | | | | - Trilokraj Tejasvi
- />Department of Dermatology, University of Michigan, Ann Arbor, MI USA
- />Ann Arbor Veterans Affairs Hospital, University of Michigan, Ann Arbor, MI USA
| | - Mrinal K Sarkar
- />Department of Dermatology, University of Michigan, Ann Arbor, MI USA
| | - Bingshan Li
- />Department of Biostatistics, Center for Statistical Genetics, School of Public Health, M4614 SPH I, University of Michigan, Box 2029, Ann Arbor, MI 48109-2029 USA
- />Department of Molecular Physiology and Biophysics, Center for Quantitative Sciences, Vanderbilt University, Nashville, TN USA
| | - Jun Ding
- />Department of Biostatistics, Center for Statistical Genetics, School of Public Health, M4614 SPH I, University of Michigan, Box 2029, Ann Arbor, MI 48109-2029 USA
- />Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - John J Voorhees
- />Department of Dermatology, University of Michigan, Ann Arbor, MI USA
| | - Hyun M Kang
- />Department of Biostatistics, Center for Statistical Genetics, School of Public Health, M4614 SPH I, University of Michigan, Box 2029, Ann Arbor, MI 48109-2029 USA
| | - Rajan P Nair
- />Department of Dermatology, University of Michigan, Ann Arbor, MI USA
| | - Arul M Chinnaiyan
- />Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI USA
- />Department of Pathology, University of Michigan Medical School, Ann Arbor, MI USA
- />Department of Urology, University of Michigan Medical School, Ann Arbor, MI USA
| | - Goncalo R Abecasis
- />Department of Biostatistics, Center for Statistical Genetics, School of Public Health, M4614 SPH I, University of Michigan, Box 2029, Ann Arbor, MI 48109-2029 USA
| | - James T Elder
- />Department of Dermatology, University of Michigan, Ann Arbor, MI USA
- />Ann Arbor Veterans Affairs Hospital, University of Michigan, Ann Arbor, MI USA
- />University of Michigan Medical School, 7412 Medical Sciences Building 1, 1301 E. Catherine, Ann Arbor, MI 48109-5675 USA
| |
Collapse
|
1078
|
Subramanian N, Torabi-Parizi P, Gottschalk RA, Germain RN, Dutta B. Network representations of immune system complexity. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 7:13-38. [PMID: 25625853 PMCID: PMC4339634 DOI: 10.1002/wsbm.1288] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 12/25/2022]
Abstract
The mammalian immune system is a dynamic multiscale system composed of a hierarchically organized set of molecular, cellular, and organismal networks that act in concert to promote effective host defense. These networks range from those involving gene regulatory and protein–protein interactions underlying intracellular signaling pathways and single‐cell responses to increasingly complex networks of in vivo cellular interaction, positioning, and migration that determine the overall immune response of an organism. Immunity is thus not the product of simple signaling events but rather nonlinear behaviors arising from dynamic, feedback‐regulated interactions among many components. One of the major goals of systems immunology is to quantitatively measure these complex multiscale spatial and temporal interactions, permitting development of computational models that can be used to predict responses to perturbation. Recent technological advances permit collection of comprehensive datasets at multiple molecular and cellular levels, while advances in network biology support representation of the relationships of components at each level as physical or functional interaction networks. The latter facilitate effective visualization of patterns and recognition of emergent properties arising from the many interactions of genes, molecules, and cells of the immune system. We illustrate the power of integrating ‘omics’ and network modeling approaches for unbiased reconstruction of signaling and transcriptional networks with a focus on applications involving the innate immune system. We further discuss future possibilities for reconstruction of increasingly complex cellular‐ and organism‐level networks and development of sophisticated computational tools for prediction of emergent immune behavior arising from the concerted action of these networks. WIREs Syst Biol Med 2015, 7:13–38. doi: 10.1002/wsbm.1288 This article is categorized under:
Analytical and Computational Methods > Computational Methods Laboratory Methods and Technologies > Macromolecular Interactions, Methods
Collapse
Affiliation(s)
- Naeha Subramanian
- Institute for Systems Biology, Seattle, WA, USA; Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
1079
|
Abstract
Systems biology and synthetic biology are emerging disciplines which are becoming increasingly utilised in several areas of bioscience. Toxicology is beginning to benefit from systems biology and we suggest in the future that is will also benefit from synthetic biology. Thus, a new era is on the horizon. This review illustrates how a suite of innovative techniques and tools can be applied to understanding complex health and toxicology issues. We review limitations confronted by the traditional computational approaches to toxicology and epidemiology research, using polycyclic aromatic hydrocarbons (PAHs) and their effects on adverse birth outcomes as an illustrative example. We introduce how systems toxicology (and their subdisciplines, genomic, proteomic, and metabolomic toxicology) will help to overcome such limitations. In particular, we discuss the advantages and disadvantages of mathematical frameworks that computationally represent biological systems. Finally, we discuss the nascent discipline of synthetic biology and highlight relevant toxicological centred applications of this technique, including improvements in personalised medicine. We conclude this review by presenting a number of opportunities and challenges that could shape the future of these rapidly evolving disciplines.
Collapse
|
1080
|
Reimand J, Wagih O, Bader GD. Evolutionary constraint and disease associations of post-translational modification sites in human genomes. PLoS Genet 2015; 11:e1004919. [PMID: 25611800 PMCID: PMC4303425 DOI: 10.1371/journal.pgen.1004919] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 11/24/2014] [Indexed: 12/14/2022] Open
Abstract
Interpreting the impact of human genome variation on phenotype is challenging. The functional effect of protein-coding variants is often predicted using sequence conservation and population frequency data, however other factors are likely relevant. We hypothesized that variants in protein post-translational modification (PTM) sites contribute to phenotype variation and disease. We analyzed fraction of rare variants and non-synonymous to synonymous variant ratio (Ka/Ks) in 7,500 human genomes and found a significant negative selection signal in PTM regions independent of six factors, including conservation, codon usage, and GC-content, that is widely distributed across tissue-specific genes and function classes. PTM regions are also enriched in known disease mutations, suggesting that PTM variation is more likely deleterious. PTM constraint also affects flanking sequence around modified residues and increases around clustered sites, indicating presence of functionally important short linear motifs. Using target site motifs of 124 kinases, we predict that at least ∼180,000 motif-breaker amino acid residues that disrupt PTM sites when substituted, and highlight kinase motifs that show specific negative selection and enrichment of disease mutations. We provide this dataset with corresponding hypothesized mechanisms as a community resource. As an example of our integrative approach, we propose that PTPN11 variants in Noonan syndrome aberrantly activate the protein by disrupting an uncharacterized cluster of phosphorylation sites. Further, as PTMs are molecular switches that are modulated by drugs, we study mutated binding sites of PTM enzymes in disease genes and define a drug-disease network containing 413 novel predicted disease-gene links.
Collapse
Affiliation(s)
- Jüri Reimand
- The Donnelly Centre, University of Toronto, Canada
- * E-mail: (JR); (GDB)
| | - Omar Wagih
- The Donnelly Centre, University of Toronto, Canada
| | - Gary D. Bader
- The Donnelly Centre, University of Toronto, Canada
- * E-mail: (JR); (GDB)
| |
Collapse
|
1081
|
Azad AKM, Lawen A, Keith JM. Prediction of signaling cross-talks contributing to acquired drug resistance in breast cancer cells by Bayesian statistical modeling. BMC SYSTEMS BIOLOGY 2015; 9:2. [PMID: 25599599 PMCID: PMC4307189 DOI: 10.1186/s12918-014-0135-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 12/11/2014] [Indexed: 01/21/2023]
Abstract
BACKGROUND Initial success of inhibitors targeting oncogenes is often followed by tumor relapse due to acquired resistance. In addition to mutations in targeted oncogenes, signaling cross-talks among pathways play a vital role in such drug inefficacy. These include activation of compensatory pathways and altered activities of key effectors in other cell survival and growth-associated pathways. RESULTS We propose a computational framework using Bayesian modeling to systematically characterize potential cross-talks among breast cancer signaling pathways. We employed a fully Bayesian approach known as the p 1-model to infer posterior probabilities of gene-pairs in networks derived from the gene expression datasets of ErbB2-positive breast cancer cell-lines (parental, lapatinib-sensitive cell-line SKBR3 and the lapatinib-resistant cell-line SKBR3-R, derived from SKBR3). Using this computational framework, we searched for cross-talks between EGFR/ErbB and other signaling pathways from Reactome, KEGG and WikiPathway databases that contribute to lapatinib resistance. We identified 104, 188 and 299 gene-pairs as putative drug-resistant cross-talks, respectively, each comprised of a gene in the EGFR/ErbB signaling pathway and a gene from another signaling pathway, that appear to be interacting in resistant cells but not in parental cells. In 168 of these (distinct) gene-pairs, both of the interacting partners are up-regulated in resistant conditions relative to parental conditions. These gene-pairs are prime candidates for novel cross-talks contributing to lapatinib resistance. They associate EGFR/ErbB signaling with six other signaling pathways: Notch, Wnt, GPCR, hedgehog, insulin receptor/IGF1R and TGF- β receptor signaling. We conducted a literature survey to validate these cross-talks, and found evidence supporting a role for many of them in contributing to drug resistance. We also analyzed an independent study of lapatinib resistance in the BT474 breast cancer cell-line and found the same signaling pathways making cross-talks with the EGFR/ErbB signaling pathway as in the primary dataset. CONCLUSIONS Our results indicate that the activation of compensatory pathways can potentially cause up-regulation of EGFR/ErbB pathway genes (counteracting the inhibiting effect of lapatinib) via signaling cross-talk. Thus, the up-regulated members of these compensatory pathways along with the members of the EGFR/ErbB signaling pathway are interesting as potential targets for designing novel anti-cancer therapeutics.
Collapse
Affiliation(s)
- A K M Azad
- School of Mathematical Science, Monash University, Wellington Road, Clayton, VIC, Australia.
| | - Alfons Lawen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Wellington Road, Clayton, VIC, Australia.
| | - Jonathan M Keith
- School of Mathematical Science, Monash University, Wellington Road, Clayton, VIC, Australia.
| |
Collapse
|
1082
|
Hutchins JRA. What's that gene (or protein)? Online resources for exploring functions of genes, transcripts, and proteins. Mol Biol Cell 2015; 25:1187-201. [PMID: 24723265 PMCID: PMC3982986 DOI: 10.1091/mbc.e13-10-0602] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The genomic era has enabled research projects that use approaches including genome-scale screens, microarray analysis, next-generation sequencing, and mass spectrometry-based proteomics to discover genes and proteins involved in biological processes. Such methods generate data sets of gene, transcript, or protein hits that researchers wish to explore to understand their properties and functions and thus their possible roles in biological systems of interest. Recent years have seen a profusion of Internet-based resources to aid this process. This review takes the viewpoint of the curious biologist wishing to explore the properties of protein-coding genes and their products, identified using genome-based technologies. Ten key questions are asked about each hit, addressing functions, phenotypes, expression, evolutionary conservation, disease association, protein structure, interactors, posttranslational modifications, and inhibitors. Answers are provided by presenting the latest publicly available resources, together with methods for hit-specific and data set-wide information retrieval, suited to any genome-based analytical technique and experimental species. The utility of these resources is demonstrated for 20 factors regulating cell proliferation. Results obtained using some of these are discussed in more depth using the p53 tumor suppressor as an example. This flexible and universally applicable approach for characterizing experimental hits helps researchers to maximize the potential of their projects for biological discovery.
Collapse
Affiliation(s)
- James R A Hutchins
- Institute of Human Genetics, Centre National de la Recherche Scientifique (CNRS), 34396 Montpellier, France
| |
Collapse
|
1083
|
Copy number variable microRNAs in schizophrenia and their neurodevelopmental gene targets. Biol Psychiatry 2015; 77:158-66. [PMID: 25034949 PMCID: PMC4464826 DOI: 10.1016/j.biopsych.2014.05.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/16/2014] [Accepted: 05/18/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) are key regulators of gene expression in the human genome and may contribute to risk for neuropsychiatric disorders. miRNAs play an acknowledged role in the strongest of genetic risk factors for schizophrenia, 22q11.2 deletions. We hypothesized that in schizophrenia there would be an enrichment of other rare copy number variants (CNVs) that overlap miRNAs. METHODS Using high-resolution genome-wide microarrays and rigorous methods, we compared the miRNA content of rare CNVs in well-characterized cohorts of schizophrenia cases (n = 420) and comparison subjects, excluding 22q11.2 CNVs. We also performed a gene-set enrichment analysis of the predicted miRNA target genes. RESULTS The schizophrenia group was enriched for the proportion of individuals with a rare CNV overlapping a miRNA (3.29-fold increase over comparison subjects, p < .0001). The presence of a rare CNV overlapping a miRNA remained a significant predictor of schizophrenia case status (p = .0072) in a multivariate logistic regression model correcting for total CNV size. In contrast, comparable analyses correcting for CNV size showed no enrichment of rare CNVs overlapping protein-coding genes. A gene-set enrichment analysis indicated that predicted target genes of recurrent CNV-overlapped miRNAs in schizophrenia may be functionally enriched for neurodevelopmental processes, including axonogenesis and neuron projection development. Predicted gene targets driving these results included CAPRIN1, NEDD4, NTRK2, PAK2, RHOA, and SYNGAP1. CONCLUSIONS These data are the first to demonstrate a genome-wide role for CNVs overlapping miRNAs in the genetic risk for schizophrenia. The results provide support for an expanded multihit model of causation, with potential implications for miRNA-based therapeutics.
Collapse
|
1084
|
Yan B, Li J, Zhang L. Identification of B cells participated in the mechanism of postmenopausal women osteoporosis using microarray analysis. Int J Clin Exp Med 2015; 8:1027-1034. [PMID: 25785089 PMCID: PMC4358544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/07/2015] [Indexed: 06/04/2023]
Abstract
To further understand the molecular mechanism of lymphocytes B cells in postmenopausal women osteoporosis. Microarray data (GSE7429) were downloaded from Gene Expression Omnibus, in which B cells were separated from the whole blood of postmenopausal women, including 10 with high bone mineral density (BMD) and 10 with low BMD. Differentially expressed genes (DEGs) between high and low BMD women were identified by Student's t-test, and P < 0.01 was used as the significant criterion. Functional enrichment analysis was performed for up- and down-regulated DEGs using KEGG, REACTOME, and Gene Ontology (GO) databases. Protein-protein interaction network (PPI) of up- and down-regulated DEGs was respectively constructed by Cytoscape software using the STRING data. Total of 169 up-regulated and 69 down-regulated DEGs were identified. Functional enrichment analysis indicated that the genes (ITPA, ATIC, UMPS, HPRT1, COX10 and COX15) might participate in metabolic pathways, MAP3K10 and MAP3K9 might participate in the activation of JNKK activity, COX10 and COX15 might involve in mitochondrial electron transport, and ATIC, UMPS and HPRT1 might involve in transferase activity. MAPK3, ITPA, ATIC, UMPS and HPRT1 with a higher degree in PPI network were identified. MAPK3, MAP3K10, MAP3K9, COX10, COX15, ATIC, UMPS and HPRT1 might participate in the pathogenesis of osteoporosis.
Collapse
|
1085
|
Rouchka EC, Chariker JH. Proceedings of the Thirteenth Annual UT- KBRIN Bioinformatics Summit 2014. BMC Bioinformatics 2015; 15 Suppl 10:I1. [PMID: 25571995 PMCID: PMC4196018 DOI: 10.1186/1471-2105-15-s10-i1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
1086
|
Sebestyén E, Zawisza M, Eyras E. Detection of recurrent alternative splicing switches in tumor samples reveals novel signatures of cancer. Nucleic Acids Res 2015; 43:1345-56. [PMID: 25578962 PMCID: PMC4330360 DOI: 10.1093/nar/gku1392] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The determination of the alternative splicing isoforms expressed in cancer is fundamental for the development of tumor-specific molecular targets for prognosis and therapy, but it is hindered by the heterogeneity of tumors and the variability across patients. We developed a new computational method, robust to biological and technical variability, which identifies significant transcript isoform changes across multiple samples. We applied this method to more than 4000 samples from the The Cancer Genome Atlas project to obtain novel splicing signatures that are predictive for nine different cancer types, and find a specific signature for basal-like breast tumors involving the tumor-driver CTNND1. Additionally, our method identifies 244 isoform switches, for which the change occurs in the most abundant transcript. Some of these switches occur in known tumor drivers, including PPARG, CCND3, RALGDS, MITF, PRDM1, ABI1 and MYH11, for which the switch implies a change in the protein product. Moreover, some of the switches cannot be described with simple splicing events. Surprisingly, isoform switches are independent of somatic mutations, except for the tumor-suppressor FBLN2 and the oncogene MYH11. Our method reveals novel signatures of cancer in terms of transcript isoforms specifically expressed in tumors, providing novel potential molecular targets for prognosis and therapy. Data and software are available at: http://dx.doi.org/10.6084/m9.figshare.1061917 and https://bitbucket.org/regulatorygenomicsupf/iso-ktsp.
Collapse
Affiliation(s)
- Endre Sebestyén
- Computational Genomics, Universitat Pompeu Fabra, Dr. Aiguader 88, E08003 Barcelona, Spain
| | - Michał Zawisza
- Universitat Politècnica de Catalunya, Jordi Girona 1-3, E08034 Barcelona, Spain
| | - Eduardo Eyras
- Computational Genomics, Universitat Pompeu Fabra, Dr. Aiguader 88, E08003 Barcelona, Spain Catalan Institution for Research and Advanced Studies, Passeig Lluís Companys 23, E08010 Barcelona, Spain
| |
Collapse
|
1087
|
Wang Y, Liu S, Hu Y, Li P, Wan JB. Current state of the art of mass spectrometry-based metabolomics studies – a review focusing on wide coverage, high throughput and easy identification. RSC Adv 2015. [DOI: 10.1039/c5ra14058g] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Metabolomics aims at the comprehensive assessment of a wide range of endogenous metabolites and attempts to identify and quantify the attractive metabolites in a given biological sample.
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory of Quality Research in Chinese Medicine
- Institute of Chinese Medical Sciences
- University of Macau
- Macao
- China
| | - Shuying Liu
- Jilin Ginseng Academy
- Changchun University of Chinese Medicine
- Changchun
- China
| | - Yuanjia Hu
- State Key Laboratory of Quality Research in Chinese Medicine
- Institute of Chinese Medical Sciences
- University of Macau
- Macao
- China
| | - Peng Li
- State Key Laboratory of Quality Research in Chinese Medicine
- Institute of Chinese Medical Sciences
- University of Macau
- Macao
- China
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine
- Institute of Chinese Medical Sciences
- University of Macau
- Macao
- China
| |
Collapse
|
1088
|
Carnielli CM, Winck FV, Paes Leme AF. Functional annotation and biological interpretation of proteomics data. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:46-54. [DOI: 10.1016/j.bbapap.2014.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/07/2014] [Accepted: 10/21/2014] [Indexed: 12/22/2022]
|
1089
|
Ogilvie LA, Wierling C, Kessler T, Lehrach H, Lange BMH. Article Commentary: Predictive Modeling of Drug Treatment in the Area of Personalized Medicine. Cancer Inform 2015. [PMID: 26692759 PMCID: PMC4671548 DOI: 10.4137/cin.s19330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Despite a growing body of knowledge on the mechanisms underlying the onset and progression of cancer, treatment success rates in oncology are at best modest. Current approaches use statistical methods that fail to embrace the inherent and expansive complexity of the tumor/patient/drug interaction. Computational modeling, in particular mechanistic modeling, has the power to resolve this complexity. Using fundamental knowledge on the interactions occurring between the components of a complex biological system, large-scale in silico models with predictive capabilities can be generated. Here, we describe how mechanistic virtual patient models, based on systematic molecular characterization of patients and their diseases, have the potential to shift the theranostic paradigm for oncology, both in the fields of personalized medicine and targeted drug development. In particular, we highlight the mechanistic modeling platform ModCell™ for individualized prediction of patient responses to treatment, emphasizing modeling techniques and avenues of application.
Collapse
Affiliation(s)
| | - Christoph Wierling
- Alacris Theranostics GmbH, Berlin, Germany
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Thomas Kessler
- Alacris Theranostics GmbH, Berlin, Germany
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Hans Lehrach
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Dahlem Centre for Genome Research and Medical Systems Biology, Berlin, Germany
| | | |
Collapse
|
1090
|
Banwait JK, Bastola DR. Contribution of bioinformatics prediction in microRNA-based cancer therapeutics. Adv Drug Deliv Rev 2015; 81:94-103. [PMID: 25450261 DOI: 10.1016/j.addr.2014.10.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 10/13/2014] [Accepted: 10/30/2014] [Indexed: 12/15/2022]
Abstract
Despite enormous efforts, cancer remains one of the most lethal diseases in the world. With the advancement of high throughput technologies massive amounts of cancer data can be accessed and analyzed. Bioinformatics provides a platform to assist biologists in developing minimally invasive biomarkers to detect cancer, and in designing effective personalized therapies to treat cancer patients. Still, the early diagnosis, prognosis, and treatment of cancer are an open challenge for the research community. MicroRNAs (miRNAs) are small non-coding RNAs that serve to regulate gene expression. The discovery of deregulated miRNAs in cancer cells and tissues has led many to investigate the use of miRNAs as potential biomarkers for early detection, and as a therapeutic agent to treat cancer. Here we describe advancements in computational approaches to predict miRNAs and their targets, and discuss the role of bioinformatics in studying miRNAs in the context of human cancer.
Collapse
Affiliation(s)
- Jasjit K Banwait
- College of Information Science and Technology, University of Nebraska at Omaha, 1110 South 67th Street, PKI 172, Omaha, NE 68106, USA.
| | - Dhundy R Bastola
- College of Information Science and Technology, University of Nebraska at Omaha, 1110 South 67th Street, PKI 172, Omaha, NE 68106, USA.
| |
Collapse
|
1091
|
Talikka M, Boue S, Schlage WK. Causal Biological Network Database: A Comprehensive Platform of Causal Biological Network Models Focused on the Pulmonary and Vascular Systems. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2015. [DOI: 10.1007/978-1-4939-2778-4_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
1092
|
Négyessy L, Györffy B, Hanics J, Bányai M, Fonta C, Bazsó F. Signal Transduction Pathways of TNAP: Molecular Network Analyses. Subcell Biochem 2015. [PMID: 26219713 DOI: 10.1007/978-94-017-7197-9_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Despite the growing body of evidence pointing on the involvement of tissue non-specific alkaline phosphatase (TNAP) in brain function and diseases like epilepsy and Alzheimer's disease, our understanding about the role of TNAP in the regulation of neurotransmission is severely limited. The aim of our study was to integrate the fragmented knowledge into a comprehensive view regarding neuronal functions of TNAP using objective tools. As a model we used the signal transduction molecular network of a pyramidal neuron after complementing with TNAP related data and performed the analysis using graph theoretic tools. The analyses show that TNAP is in the crossroad of numerous pathways and therefore is one of the key players of the neuronal signal transduction network. Through many of its connections, most notably with molecules of the purinergic system, TNAP serves as a controller by funnelling signal flow towards a subset of molecules. TNAP also appears as the source of signal to be spread via interactions with molecules involved among others in neurodegeneration. Cluster analyses identified TNAP as part of the second messenger signalling cascade. However, TNAP also forms connections with other functional groups involved in neuronal signal transduction. The results indicate the distinct ways of involvement of TNAP in multiple neuronal functions and diseases.
Collapse
Affiliation(s)
- László Négyessy
- Theoretical Neuroscience and Complex Systems Research Group, Wigner Research Center for Physics, Budapest, Hungary,
| | | | | | | | | | | |
Collapse
|
1093
|
Abstract
Metabolites as an end product of metabolism possess a wealth of information about altered metabolic control and homeostasis that is dependent on numerous variables including age, sex, and environment. Studying significant changes in the metabolite patterns has been recognized as a tool to understand crucial aspects in drug development like drug efficacy and toxicity. The inclusion of metabonomics into the OMICS study platform brings us closer to define the phenotype and allows us to look at alternatives to improve the diagnosis of diseases. Advancements in the analytical strategies and statistical tools used to study metabonomics allow us to prevent drug failures at early stages of drug development and reduce financial losses during expensive phase II and III clinical trials. This chapter introduces metabonomics along with the instruments used in the study; in addition relevant examples of the usage of metabonomics in the drug development process are discussed along with an emphasis on future directions and the challenges it faces.
Collapse
Affiliation(s)
- Pranov Ramana
- Pharmaceutical Analysis, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, O&N2 PB 923, Herestraat 49, 3000, Leuven, Belgium
| | | | | | | |
Collapse
|
1094
|
Liu Y, Wei Q, Yu G, Gai W, Li Y, Chen X. DCDB 2.0: a major update of the drug combination database. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2014; 2014:bau124. [PMID: 25539768 PMCID: PMC4275564 DOI: 10.1093/database/bau124] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Experience in clinical practice and research in systems pharmacology suggested the limitations of the current one-drug-one-target paradigm in new drug discovery. Single-target drugs may not always produce desired physiological effects on the entire biological system, even if they have successfully regulated the activities of their designated targets. On the other hand, multicomponent therapy, in which two or more agents simultaneously interact with multiple targets, has attracted growing attention. Many drug combinations consisting of multiple agents have already entered clinical practice, especially in treating complex and refractory diseases. Drug combination database (DCDB), launched in 2010, is the first available database that collects and organizes information on drug combinations, with an aim to facilitate systems-oriented new drug discovery. Here, we report the second major release of DCDB (Version 2.0), which includes 866 new drug combinations (1363 in total), consisting of 904 distinctive components. These drug combinations are curated from ∼140,000 clinical studies and the food and drug administration (FDA) electronic orange book. In this update, DCDB collects 237 unsuccessful drug combinations, which may provide a contrast for systematic discovery of the patterns in successful drug combinations. Database URL: http://www.cls.zju.edu.cn/dcdb/
Collapse
Affiliation(s)
- Yanbin Liu
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Qiang Wei
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Guisheng Yu
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Wanxia Gai
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yongquan Li
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Xin Chen
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| |
Collapse
|
1095
|
Hornbeck PV, Zhang B, Murray B, Kornhauser JM, Latham V, Skrzypek E. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res 2014; 43:D512-20. [PMID: 25514926 PMCID: PMC4383998 DOI: 10.1093/nar/gku1267] [Citation(s) in RCA: 2175] [Impact Index Per Article: 217.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PhosphoSitePlus® (PSP, http://www.phosphosite.org/), a knowledgebase dedicated to mammalian post-translational modifications (PTMs), contains over 330 000 non-redundant PTMs, including phospho, acetyl, ubiquityl and methyl groups. Over 95% of the sites are from mass spectrometry (MS) experiments. In order to improve data reliability, early MS data have been reanalyzed, applying a common standard of analysis across over 1 000 000 spectra. Site assignments with P > 0.05 were filtered out. Two new downloads are available from PSP. The ‘Regulatory sites’ dataset includes curated information about modification sites that regulate downstream cellular processes, molecular functions and protein-protein interactions. The ‘PTMVar’ dataset, an intersect of missense mutations and PTMs from PSP, identifies over 25 000 PTMVars (PTMs Impacted by Variants) that can rewire signaling pathways. The PTMVar data include missense mutations from UniPROTKB, TCGA and other sources that cause over 2000 diseases or syndromes (MIM) and polymorphisms, or are associated with hundreds of cancers. PTMVars include 18 548 phosphorlyation sites, 3412 ubiquitylation sites, 2316 acetylation sites, 685 methylation sites and 245 succinylation sites.
Collapse
Affiliation(s)
| | - Bin Zhang
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA 01923, USA
| | - Beth Murray
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA 01923, USA
| | | | - Vaughan Latham
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA 01923, USA
| | | |
Collapse
|
1096
|
Iourov IY, Vorsanova SG, Yurov YB. In silico molecular cytogenetics: a bioinformatic approach to prioritization of candidate genes and copy number variations for basic and clinical genome research. Mol Cytogenet 2014; 7:98. [PMID: 25525469 PMCID: PMC4269961 DOI: 10.1186/s13039-014-0098-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 12/02/2014] [Indexed: 01/08/2023] Open
Abstract
Background The availability of multiple in silico tools for prioritizing genetic variants widens the possibilities for converting genomic data into biological knowledge. However, in molecular cytogenetics, bioinformatic analyses are generally limited to result visualization or database mining for finding similar cytogenetic data. Obviously, the potential of bioinformatics might go beyond these applications. On the other hand, the requirements for performing successful in silico analyses (i.e. deep knowledge of computer science, statistics etc.) can hinder the implementation of bioinformatics in clinical and basic molecular cytogenetic research. Here, we propose a bioinformatic approach to prioritization of genomic variations that is able to solve these problems. Results Selecting gene expression as an initial criterion, we have proposed a bioinformatic approach combining filtering and ranking prioritization strategies, which includes analyzing metabolome and interactome data on proteins encoded by candidate genes. To finalize the prioritization of genetic variants, genomic, epigenomic, interactomic and metabolomic data fusion has been made. Structural abnormalities and aneuploidy revealed by array CGH and FISH have been evaluated to test the approach through determining genotype-phenotype correlations, which have been found similar to those of previous studies. Additionally, we have been able to prioritize copy number variations (CNV) (i.e. differentiate between benign CNV and CNV with phenotypic outcome). Finally, the approach has been applied to prioritize genetic variants in cases of somatic mosaicism (including tissue-specific mosaicism). Conclusions In order to provide for an in silico evaluation of molecular cytogenetic data, we have proposed a bioinformatic approach to prioritization of candidate genes and CNV. While having the disadvantage of possible unavailability of gene expression data or lack of expression variability between genes of interest, the approach provides several advantages. These are (i) the versatility due to independence from specific databases/tools or software, (ii) relative algorithm simplicity (possibility to avoid sophisticated computational/statistical methodology) and (iii) applicability to molecular cytogenetic data because of the chromosome-centric nature. In conclusion, the approach is able to become useful for increasing the yield of molecular cytogenetic techniques.
Collapse
Affiliation(s)
- Ivan Y Iourov
- Mental Health Research Center, Russian Academy of Medical Sciences, 117152 Moscow, Russia ; Russian National Research Medical University named after N.I. Pirogov, Separated Structural Unit "Clinical Research Institute of Pediatrics", Ministry of Health of Russian Federation, 125412 Moscow, Russia ; Department of Medical Genetics, Russian Medical Academy of Postgraduate Education, Moscow, 123995 Russia
| | - Svetlana G Vorsanova
- Mental Health Research Center, Russian Academy of Medical Sciences, 117152 Moscow, Russia ; Russian National Research Medical University named after N.I. Pirogov, Separated Structural Unit "Clinical Research Institute of Pediatrics", Ministry of Health of Russian Federation, 125412 Moscow, Russia
| | - Yuri B Yurov
- Mental Health Research Center, Russian Academy of Medical Sciences, 117152 Moscow, Russia ; Russian National Research Medical University named after N.I. Pirogov, Separated Structural Unit "Clinical Research Institute of Pediatrics", Ministry of Health of Russian Federation, 125412 Moscow, Russia
| |
Collapse
|
1097
|
Kunz M, Xiao K, Liang C, Viereck J, Pachel C, Frantz S, Thum T, Dandekar T. Bioinformatics of cardiovascular miRNA biology. J Mol Cell Cardiol 2014; 89:3-10. [PMID: 25486579 DOI: 10.1016/j.yjmcc.2014.11.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/05/2014] [Accepted: 11/29/2014] [Indexed: 12/16/2022]
Abstract
MicroRNAs (miRNAs) are small ~22 nucleotide non-coding RNAs and are highly conserved among species. Moreover, miRNAs regulate gene expression of a large number of genes associated with important biological functions and signaling pathways. Recently, several miRNAs have been found to be associated with cardiovascular diseases. Thus, investigating the complex regulatory effect of miRNAs may lead to a better understanding of their functional role in the heart. To achieve this, bioinformatics approaches have to be coupled with validation and screening experiments to understand the complex interactions of miRNAs with the genome. This will boost the subsequent development of diagnostic markers and our understanding of the physiological and therapeutic role of miRNAs in cardiac remodeling. In this review, we focus on and explain different bioinformatics strategies and algorithms for the identification and analysis of miRNAs and their regulatory elements to better understand cardiac miRNA biology. Starting with the biogenesis of miRNAs, we present approaches such as LocARNA and miRBase for combining sequence and structure analysis including phylogenetic comparisons as well as detailed analysis of RNA folding patterns, functional target prediction, signaling pathway as well as functional analysis. We also show how far bioinformatics helps to tackle the unprecedented level of complexity and systemic effects by miRNA, underlining the strong therapeutic potential of miRNA and miRNA target structures in cardiovascular disease. In addition, we discuss drawbacks and limitations of bioinformatics algorithms and the necessity of experimental approaches for miRNA target identification. This article is part of a Special Issue entitled 'Non-coding RNAs'.
Collapse
Affiliation(s)
- Meik Kunz
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Würzburg, Germany; Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Ke Xiao
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany; Plant Breeding Institute, Christian-Albrechts-University of Kiel, Olshausenstr. 40, 24098 Kiel, Germany
| | - Chunguang Liang
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Würzburg, Germany
| | - Janika Viereck
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Christina Pachel
- Department of Internal Medicine I, University Hospital Würzburg, Germany and Comprehensive Heart Failure Center, University of Würzburg, Germany
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Germany and Comprehensive Heart Failure Center, University of Würzburg, Germany
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany; Excellence Cluster REBIRTH, Hannover Medical School, Hannover, Germany; National Heart and Lung Institute, Imperial College London, London, UK
| | - Thomas Dandekar
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Würzburg, Germany; EMBL Heidelberg, BioComputing Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany.
| |
Collapse
|
1098
|
Tsoi LC, Elder JT, Abecasis GR. Graphical algorithm for integration of genetic and biological data: proof of principle using psoriasis as a model. ACTA ACUST UNITED AC 2014; 31:1243-9. [PMID: 25480373 DOI: 10.1093/bioinformatics/btu799] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/26/2014] [Indexed: 01/17/2023]
Abstract
MOTIVATION Pathway analysis to reveal biological mechanisms for results from genetic association studies have great potential to better understand complex traits with major human disease impact. However, current approaches have not been optimized to maximize statistical power to identify enriched functions/pathways, especially when the genetic data derives from studies using platforms (e.g. Immunochip and Metabochip) customized to have pre-selected markers from previously identified top-rank loci. We present here a novel approach, called Minimum distance-based Enrichment Analysis for Genetic Association (MEAGA), with the potential to address both of these important concerns. RESULTS MEAGA performs enrichment analysis using graphical algorithms to identify sub-graphs among genes and measure their closeness in interaction database. It also incorporates a statistic summarizing the numbers and total distances of the sub-graphs, depicting the overlap between observed genetic signals and defined function/pathway gene-sets. MEAGA uses sampling technique to approximate empirical and multiple testing-corrected P-values. We show in simulation studies that MEAGA is more powerful compared to count-based strategies in identifying disease-associated functions/pathways, and the increase in power is influenced by the shortest distances among associated genes in the interactome. We applied MEAGA to the results of a meta-analysis of psoriasis using Immunochip datasets, and showed that associated genes are significantly enriched in immune-related functions and closer with each other in the protein-protein interaction network. AVAILABILITY AND IMPLEMENTATION http://genome.sph.umich.edu/wiki/MEAGA CONTACT: : tsoi.teen@gmail.com or goncalo@umich.edu SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Lam C Tsoi
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA, Department of Dermatology, University of Michigan, Ann Arbor, MI, USA, and Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA
| | - James T Elder
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA, Department of Dermatology, University of Michigan, Ann Arbor, MI, USA, and Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA, Department of Dermatology, University of Michigan, Ann Arbor, MI, USA, and Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA
| | - Goncalo R Abecasis
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA, Department of Dermatology, University of Michigan, Ann Arbor, MI, USA, and Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA
| |
Collapse
|
1099
|
Cakır T, Khatibipour MJ. Metabolic network discovery by top-down and bottom-up approaches and paths for reconciliation. Front Bioeng Biotechnol 2014; 2:62. [PMID: 25520953 PMCID: PMC4253960 DOI: 10.3389/fbioe.2014.00062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/14/2014] [Indexed: 11/13/2022] Open
Abstract
The primary focus in the network-centric analysis of cellular metabolism by systems biology approaches is to identify the active metabolic network for the condition of interest. Two major approaches are available for the discovery of the condition-specific metabolic networks. One approach starts from genome-scale metabolic networks, which cover all possible reactions known to occur in the related organism in a condition-independent manner, and applies methods such as the optimization-based Flux-Balance Analysis to elucidate the active network. The other approach starts from the condition-specific metabolome data, and processes the data with statistical or optimization-based methods to extract information content of the data such that the active network is inferred. These approaches, termed bottom-up and top-down, respectively, are currently employed independently. However, considering that both approaches have the same goal, they can both benefit from each other paving the way for the novel integrative analysis methods of metabolome data- and flux-analysis approaches in the post-genomic era. This study reviews the strengths of constraint-based analysis and network inference methods reported in the metabolic systems biology field; then elaborates on the potential paths to reconcile the two approaches to shed better light on how the metabolism functions.
Collapse
Affiliation(s)
- Tunahan Cakır
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University (formerly known as Gebze Institute of Technology) , Gebze , Turkey
| | - Mohammad Jafar Khatibipour
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University (formerly known as Gebze Institute of Technology) , Gebze , Turkey ; Department of Chemical Engineering, Gebze Technical University (formerly known as Gebze Institute of Technology) , Gebze , Turkey
| |
Collapse
|
1100
|
Mosca E, Alfieri R, Milanesi L. Diffusion of information throughout the host interactome reveals gene expression variations in network proximity to target proteins of hepatitis C virus. PLoS One 2014; 9:e113660. [PMID: 25461596 PMCID: PMC4251971 DOI: 10.1371/journal.pone.0113660] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/27/2014] [Indexed: 12/22/2022] Open
Abstract
Hepatitis C virus infection is one of the most common and chronic in the world, and hepatitis associated with HCV infection is a major risk factor for the development of cirrhosis and hepatocellular carcinoma (HCC). The rapidly growing number of viral-host and host protein-protein interactions is enabling more and more reliable network-based analyses of viral infection supported by omics data. The study of molecular interaction networks helps to elucidate the mechanistic pathways linking HCV molecular activities and the host response that modulates the stepwise hepatocarcinogenic process from preneoplastic lesions (cirrhosis and dysplasia) to HCC. Simulating the impact of HCV-host molecular interactions throughout the host protein-protein interaction (PPI) network, we ranked the host proteins in relation to their network proximity to viral targets. We observed that the set of proteins in the neighborhood of HCV targets in the host interactome is enriched in key players of the host response to HCV infection. In opposition to HCV targets, subnetworks of proteins in network proximity to HCV targets are significantly enriched in proteins reported as differentially expressed in preneoplastic and neoplastic liver samples by two independent studies. Using multi-objective optimization, we extracted subnetworks that are simultaneously “guilt-by-association” with HCV proteins and enriched in proteins differentially expressed. These subnetworks contain established, recently proposed and novel candidate proteins for the regulation of the mechanisms of liver cells response to chronic HCV infection.
Collapse
Affiliation(s)
- Ettore Mosca
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, Italy
- * E-mail:
| | - Roberta Alfieri
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, Italy
| |
Collapse
|