51
|
Bansal A, Srivastava PA, Singh TR. An integrative approach to develop computational pipeline for drug-target interaction network analysis. Sci Rep 2018; 8:10238. [PMID: 29980766 PMCID: PMC6035197 DOI: 10.1038/s41598-018-28577-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/26/2018] [Indexed: 11/25/2022] Open
Abstract
Understanding the general principles governing the functioning of biological networks is a major challenge of the current era. Functionality of biological networks can be observed from drug and target interaction perspective. All possible modes of operations of biological networks are confined by the interaction analysis. Several of the existing approaches in this direction, however, are data-driven and thus lack potential to be generalized and extrapolated to different species. In this paper, we demonstrate a systems pharmacology pipeline and discuss how the network theory, along with gene ontology (GO) analysis, co-expression analysis, module re-construction, pathway mapping and structure level analysis can be used to decipher important properties of biological networks with the aim to propose lead molecule for the therapeutic interventions of various diseases.
Collapse
Affiliation(s)
- Ankush Bansal
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, 173234, Solan, HP, India
| | - Pulkit Anupam Srivastava
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, 173234, Solan, HP, India
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, 173234, Solan, HP, India.
| |
Collapse
|
52
|
Aguirre-Plans J, Piñero J, Menche J, Sanz F, Furlong LI, Schmidt HHHW, Oliva B, Guney E. Proximal Pathway Enrichment Analysis for Targeting Comorbid Diseases via Network Endopharmacology. Pharmaceuticals (Basel) 2018; 11:E61. [PMID: 29932108 PMCID: PMC6160959 DOI: 10.3390/ph11030061] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 01/13/2023] Open
Abstract
The past decades have witnessed a paradigm shift from the traditional drug discovery shaped around the idea of “one target, one disease” to polypharmacology (multiple targets, one disease). Given the lack of clear-cut boundaries across disease (endo)phenotypes and genetic heterogeneity across patients, a natural extension to the current polypharmacology paradigm is to target common biological pathways involved in diseases via endopharmacology (multiple targets, multiple diseases). In this study, we present proximal pathway enrichment analysis (PxEA) for pinpointing drugs that target common disease pathways towards network endopharmacology. PxEA uses the topology information of the network of interactions between disease genes, pathway genes, drug targets and other proteins to rank drugs by their interactome-based proximity to pathways shared across multiple diseases, providing unprecedented drug repurposing opportunities. Using PxEA, we show that many drugs indicated for autoimmune disorders are not necessarily specific to the condition of interest, but rather target the common biological pathways across these diseases. Finally, we provide high scoring drug repurposing candidates that can target common mechanisms involved in type 2 diabetes and Alzheimer’s disease, two conditions that have recently gained attention due to the increased comorbidity among patients.
Collapse
Affiliation(s)
- Joaquim Aguirre-Plans
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Janet Piñero
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Jörg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, A-1090 Vienna, Austria.
| | - Ferran Sanz
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Laura I Furlong
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, CARIM, FHML, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands.
| | - Baldo Oliva
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Emre Guney
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
- Department of Pharmacology and Personalised Medicine, CARIM, FHML, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
53
|
Barouki R, Melén E, Herceg Z, Beckers J, Chen J, Karagas M, Puga A, Xia Y, Chadwick L, Yan W, Audouze K, Slama R, Heindel J, Grandjean P, Kawamoto T, Nohara K. Epigenetics as a mechanism linking developmental exposures to long-term toxicity. ENVIRONMENT INTERNATIONAL 2018; 114:77-86. [PMID: 29499450 PMCID: PMC5899930 DOI: 10.1016/j.envint.2018.02.014] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/13/2018] [Accepted: 02/08/2018] [Indexed: 05/17/2023]
Abstract
A variety of experimental and epidemiological studies lend support to the Developmental Origin of Health and Disease (DOHaD) concept. Yet, the actual mechanisms accounting for mid- and long-term effects of early-life exposures remain unclear. Epigenetic alterations such as changes in DNA methylation, histone modifications and the expression of certain RNAs have been suggested as possible mediators of long-term health effects of environmental stressors. This report captures discussions and conclusions debated during the last Prenatal Programming and Toxicity meeting held in Japan. Its first aim is to propose a number of criteria that are critical to support the primary contribution of epigenetics in DOHaD and intergenerational transmission of environmental stressors effects. The main criteria are the full characterization of the stressors, the actual window of exposure, the target tissue and function, the specificity of the epigenetic changes and the biological plausibility of the linkage between those changes and health outcomes. The second aim is to discuss long-term effects of a number of stressors such as smoking, air pollution and endocrine disruptors in order to identify the arguments supporting the involvement of an epigenetic mechanism. Based on the developed criteria, missing evidence and suggestions for future research will be identified. The third aim is to critically analyze the evidence supporting the involvement of epigenetic mechanisms in intergenerational and transgenerational effects of environmental exposure and to particularly discuss the role of placenta and sperm. While the article is not a systematic review and is not meant to be exhaustive, it critically assesses the contribution of epigenetics in the long-term effects of environmental exposures as well as provides insight for future research.
Collapse
Affiliation(s)
- R Barouki
- INSERM UMR-S 1124, Université Paris Descartes, Paris, France; Service de Biochimie Métabolomique et Protéomique, Hôpital Necker Enfants Malades, AP-HP, Paris, France.
| | - E Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Sachs' Children and Youth Hospital, and Centre for Occupational and Environmental Medicine, Stockholm County Council, Sweden
| | - Z Herceg
- Epigenetics Group, International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, F-69008 Lyon, France
| | - J Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, 85764 Neuherberg, Germany; Technische Universität München, Experimental Genetics, 85354 Freising, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - J Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - M Karagas
- Department of Epidemiology, Children's Environmental Health and Disease Prevention Research Center at Dartmouth, Hanover, NH, USA
| | - A Puga
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Y Xia
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | | | - W Yan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV 89557, USA MS575; Department of Biology, University of Nevada, Reno, 1664 North Virginia Street, Reno, NV 89557, USA
| | - K Audouze
- INSERM UMR-S973, Molécules Thérapeutiques in silico, University of Paris Diderot, Paris, France
| | - R Slama
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, Grenoble, France
| | - J Heindel
- Program in Endocrine Disruption Strategies, Commonweal, Bolinas, CA, USA
| | - P Grandjean
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - T Kawamoto
- Department of Environmental Health, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - K Nohara
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba 305-8506, Japan
| |
Collapse
|
54
|
Woidy M, Muntau AC, Gersting SW. Inborn errors of metabolism and the human interactome: a systems medicine approach. J Inherit Metab Dis 2018; 41:285-296. [PMID: 29404805 PMCID: PMC5959957 DOI: 10.1007/s10545-018-0140-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 01/01/2018] [Accepted: 01/10/2018] [Indexed: 12/14/2022]
Abstract
The group of inborn errors of metabolism (IEM) displays a marked heterogeneity and IEM can affect virtually all functions and organs of the human organism; however, IEM share that their associated proteins function in metabolism. Most proteins carry out cellular functions by interacting with other proteins, and thus are organized in biological networks. Therefore, diseases are rarely the consequence of single gene mutations but of the perturbations caused in the related cellular network. Systematic approaches that integrate multi-omics and database information into biological networks have successfully expanded our knowledge of complex disorders but network-based strategies have been rarely applied to study IEM. We analyzed IEM on a proteome scale and found that IEM-associated proteins are organized as a network of linked modules within the human interactome of protein interactions, the IEM interactome. Certain IEM disease groups formed self-contained disease modules, which were highly interlinked. On the other hand, we observed disease modules consisting of proteins from many different disease groups in the IEM interactome. Moreover, we explored the overlap between IEM and non-IEM disease genes and applied network medicine approaches to investigate shared biological pathways, clinical signs and symptoms, and links to drug targets. The provided resources may help to elucidate the molecular mechanisms underlying new IEM, to uncover the significance of disease-associated mutations, to identify new biomarkers, and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Mathias Woidy
- University Children's Hospital, University Medical Center Hamburg Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Ania C Muntau
- University Children's Hospital, University Medical Center Hamburg Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Søren W Gersting
- Department of Molecular Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstrasse 4, 80336, Munich, Germany.
| |
Collapse
|
55
|
Abstract
Precision medicine is an integrative approach to cardiovascular disease prevention and treatment that considers an individual's genetics, lifestyle, and exposures as determinants of their cardiovascular health and disease phenotypes. This focus overcomes the limitations of reductionism in medicine, which presumes that all patients with the same signs of disease share a common pathophenotype and, therefore, should be treated similarly. Precision medicine incorporates standard clinical and health record data with advanced panomics (ie, transcriptomics, epigenomics, proteomics, metabolomics, and microbiomics) for deep phenotyping. These phenotypic data can then be analyzed within the framework of molecular interaction (interactome) networks to uncover previously unrecognized disease phenotypes and relationships between diseases, and to select pharmacotherapeutics or identify potential protein-drug or drug-drug interactions. In this review, we discuss the current spectrum of cardiovascular health and disease, population averages and the response of extreme phenotypes to interventions, and population-based versus high-risk treatment strategies as a pretext to understanding a precision medicine approach to cardiovascular disease prevention and therapeutic interventions. We also consider the search for resilience and Mendelian disease genes and argue against the theory of a single causal gene/gene product as a mediator of the cardiovascular disease phenotype, as well as an Erlichian magic bullet to solve cardiovascular disease. Finally, we detail the importance of deep phenotyping and interactome networks and the use of this information for rational polypharmacy. These topics highlight the urgent need for precise phenotyping to advance precision medicine as a strategy to improve cardiovascular health and prevent disease.
Collapse
Affiliation(s)
- Jane A Leopold
- From the Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Joseph Loscalzo
- From the Brigham and Women's Hospital and Harvard Medical School, Boston, MA.
| |
Collapse
|
56
|
Parolo S, Marchetti L, Lauria M, Misselbeck K, Scott-Boyer MP, Caberlotto L, Priami C. Combined use of protein biomarkers and network analysis unveils deregulated regulatory circuits in Duchenne muscular dystrophy. PLoS One 2018. [PMID: 29529088 PMCID: PMC5846794 DOI: 10.1371/journal.pone.0194225] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although the genetic basis of Duchenne muscular dystrophy has been known for almost thirty years, the cellular and molecular mechanisms characterizing the disease are not completely understood and an efficacious treatment remains to be developed. In this study we analyzed proteomics data obtained with the SomaLogic technology from blood serum of a cohort of patients and matched healthy subjects. We developed a workflow based on biomarker identification and network-based pathway analysis that allowed us to describe different deregulated pathways. In addition to muscle-related functions, we identified other biological processes such as apoptosis, signaling in the immune system and neurotrophin signaling as significantly modulated in patients compared with controls. Moreover, our network-based analysis identified the involvement of FoxO transcription factors as putative regulators of different pathways. On the whole, this study provided a global view of the molecular processes involved in Duchenne muscular dystrophy that are decipherable from serum proteome.
Collapse
Affiliation(s)
- Silvia Parolo
- The Microsoft Research—University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto (TN), Italy
- * E-mail:
| | - Luca Marchetti
- The Microsoft Research—University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto (TN), Italy
| | - Mario Lauria
- The Microsoft Research—University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto (TN), Italy
- Department of Mathematics, University of Trento, Povo (TN), Italy
| | - Karla Misselbeck
- The Microsoft Research—University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto (TN), Italy
- Department of Mathematics, University of Trento, Povo (TN), Italy
| | - Marie-Pier Scott-Boyer
- The Microsoft Research—University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto (TN), Italy
| | - Laura Caberlotto
- The Microsoft Research—University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto (TN), Italy
| | - Corrado Priami
- The Microsoft Research—University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto (TN), Italy
- Department of Computer Science, University of Pisa, Pisa (PI), Italy
| |
Collapse
|
57
|
A diseasome cluster-based drug repurposing of soluble guanylate cyclase activators from smooth muscle relaxation to direct neuroprotection. NPJ Syst Biol Appl 2018; 4:8. [PMID: 29423274 PMCID: PMC5799370 DOI: 10.1038/s41540-017-0039-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 12/24/2022] Open
Abstract
Network medicine utilizes common genetic origins, markers and co-morbidities to uncover mechanistic links between diseases. These links can be summarized in the diseasome, a comprehensive network of disease–disease relationships and clusters. The diseasome has been influential during the past decade, although most of its links are not followed up experimentally. Here, we investigate a high prevalence unmet medical need cluster of disease phenotypes linked to cyclic GMP. Hitherto, the central cGMP-forming enzyme, soluble guanylate cyclase (sGC), has been targeted pharmacologically exclusively for smooth muscle modulation in cardiology and pulmonology. Here, we examine the disease associations of sGC in a non-hypothesis based manner in order to identify possibly previously unrecognized clinical indications. Surprisingly, we find that sGC, is closest linked to neurological disorders, an application that has so far not been explored clinically. Indeed, when investigating the neurological indication of this cluster with the highest unmet medical need, ischemic stroke, pre-clinically we find that sGC activity is virtually absent post-stroke. Conversely, a heme-free form of sGC, apo-sGC, was now the predominant isoform suggesting it may be a mechanism-based target in stroke. Indeed, this repurposing hypothesis could be validated experimentally in vivo as specific activators of apo-sGC were directly neuroprotective, reduced infarct size and increased survival. Thus, common mechanism clusters of the diseasome allow direct drug repurposing across previously unrelated disease phenotypes redefining them in a mechanism-based manner. Specifically, our example of repurposing apo-sGC activators for ischemic stroke should be urgently validated clinically as a possible first-in-class neuroprotective therapy. Systems medicine utilizes common genetic origins and co-morbidities to uncover mechanistic links between diseases, which are summarized in the diseasome. Shared pathomechanisms may also allow for drug repurposing within these disease clusters. Here, Schmidt and co-workers show indeed that, based on this principle, a cardio-pulmonary drug can be surprisingly repurposed for a previously not recognised application as a direct neuroprotectant. They find that the cyclic GMP forming soluble guanylate cyclase becomes dysfunctional upon stroke but regains catalytic activity in the presence of specific activator compounds. This new mechanism-based therapy should be urgently validated clinically as a possible first-in-class treatment in stroke.
Collapse
|
58
|
Cheng F, Loscalzo J. Pulmonary Comorbidity in Lung Cancer. Trends Mol Med 2018; 24:239-241. [PMID: 29398402 DOI: 10.1016/j.molmed.2018.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 01/17/2018] [Indexed: 01/20/2023]
Abstract
Pulmonary hypertension (PH) is caused by many disorders that affect the pulmonary vasculature. A recent study has provided evidence that pulmonary vascular remodeling and PH can be observed in lung cancer, and this may be associated with tumor cell-immune cell inflammatory crosstalk. These findings highlight the pressing need to understand better and manage pulmonary vascular comorbidities in lung cancer.
Collapse
Affiliation(s)
- Feixiong Cheng
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA 02115, USA; Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
59
|
Greene JA, Loscalzo J. Putting the Patient Back Together - Social Medicine, Network Medicine, and the Limits of Reductionism. N Engl J Med 2017; 377:2493-2499. [PMID: 29262277 DOI: 10.1056/nejmms1706744] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Jeremy A Greene
- From the Departments of Medicine and the History of Medicine and the Center for Medical Humanities and Social Medicine, Johns Hopkins University School of Medicine, Baltimore (J.A.G.); and the Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston (J.L.)
| | - Joseph Loscalzo
- From the Departments of Medicine and the History of Medicine and the Center for Medical Humanities and Social Medicine, Johns Hopkins University School of Medicine, Baltimore (J.A.G.); and the Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston (J.L.)
| |
Collapse
|
60
|
Khamina K, Lercher A, Caldera M, Schliehe C, Vilagos B, Sahin M, Kosack L, Bhattacharya A, Májek P, Stukalov A, Sacco R, James LC, Pinschewer DD, Bennett KL, Menche J, Bergthaler A. Characterization of host proteins interacting with the lymphocytic choriomeningitis virus L protein. PLoS Pathog 2017; 13:e1006758. [PMID: 29261807 PMCID: PMC5738113 DOI: 10.1371/journal.ppat.1006758] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/17/2017] [Indexed: 01/10/2023] Open
Abstract
RNA-dependent RNA polymerases (RdRps) play a key role in the life cycle of RNA viruses and impact their immunobiology. The arenavirus lymphocytic choriomeningitis virus (LCMV) strain Clone 13 provides a benchmark model for studying chronic infection. A major genetic determinant for its ability to persist maps to a single amino acid exchange in the viral L protein, which exhibits RdRp activity, yet its functional consequences remain elusive. To unravel the L protein interactions with the host proteome, we engineered infectious L protein-tagged LCMV virions by reverse genetics. A subsequent mass-spectrometric analysis of L protein pulldowns from infected human cells revealed a comprehensive network of interacting host proteins. The obtained LCMV L protein interactome was bioinformatically integrated with known host protein interactors of RdRps from other RNA viruses, emphasizing interconnected modules of human proteins. Functional characterization of selected interactors highlighted proviral (DDX3X) as well as antiviral (NKRF, TRIM21) host factors. To corroborate these findings, we infected Trim21-/- mice with LCMV and found impaired virus control in chronic infection. These results provide insights into the complex interactions of the arenavirus LCMV and other viral RdRps with the host proteome and contribute to a better molecular understanding of how chronic viruses interact with their host. RNA-dependent RNA-polymerases (RdRps) play a key role in the life cycle of RNA viruses. They interact with cellular proteins during replication and transcription processes and impact the immunobiology of viral infections. This study characterized the host protein interactome of the RdRp-containing L protein of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV). Several L protein interactors with proviral and antiviral effects were identified in vitro, and mice lacking the identified L protein interactor TRIM21 exhibited impaired control of chronic LCMV infection. Integration of the L protein interactomes with known RdRp interactomes from other RNA viruses highlighted common and virus-specific strategies to interact with the host proteome, which may indicate novel avenues for antiviral interventions.
Collapse
Affiliation(s)
- Kseniya Khamina
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Alexander Lercher
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Michael Caldera
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Christopher Schliehe
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Bojan Vilagos
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Mehmet Sahin
- University of Basel, Department of Biomedicine–Haus Petersplatz, Division of Experimental Virology, Basel, Switzerland
| | - Lindsay Kosack
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Anannya Bhattacharya
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Peter Májek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Alexey Stukalov
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Roberto Sacco
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Leo C. James
- Division of Protein and Nucleic Acid Chemistry, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Daniel D. Pinschewer
- University of Basel, Department of Biomedicine–Haus Petersplatz, Division of Experimental Virology, Basel, Switzerland
| | - Keiryn L. Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Jörg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
- * E-mail:
| |
Collapse
|
61
|
de Souza HSP, Fiocchi C, Iliopoulos D. The IBD interactome: an integrated view of aetiology, pathogenesis and therapy. Nat Rev Gastroenterol Hepatol 2017; 14:739-749. [PMID: 28831186 DOI: 10.1038/nrgastro.2017.110] [Citation(s) in RCA: 303] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Crohn's disease and ulcerative colitis are prototypical complex diseases characterized by chronic and heterogeneous manifestations, induced by interacting environmental, genomic, microbial and immunological factors. These interactions result in an overwhelming complexity that cannot be tackled by studying the totality of each pathological component (an '-ome') in isolation without consideration of the interaction among all relevant -omes that yield an overall 'network effect'. The outcome of this effect is the 'IBD interactome', defined as a disease network in which dysregulation of individual -omes causes intestinal inflammation mediated by dysfunctional molecular modules. To define the IBD interactome, new concepts and tools are needed to implement a systems approach; an unbiased data-driven integration strategy that reveals key players of the system, pinpoints the central drivers of inflammation and enables development of targeted therapies. Powerful bioinformatics tools able to query and integrate multiple -omes are available, enabling the integration of genomic, epigenomic, transcriptomic, proteomic, metabolomic and microbiome information to build a comprehensive molecular map of IBD. This approach will enable identification of IBD molecular subtypes, correlations with clinical phenotypes and elucidation of the central hubs of the IBD interactome that will aid discovery of compounds that can specifically target the hubs that control the disease.
Collapse
Affiliation(s)
- Heitor S P de Souza
- Department of Gastroenterology & Multidisciplinary Research Laboratory, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Claudio Fiocchi
- Department of Pathobiology, Lerner Research Institute, Department of Gastroenterology and Hepatology, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Dimitrios Iliopoulos
- Center for Systems Biomedicine, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
62
|
Costa MD, Davis RB, Goldberger AL. Heart Rate Fragmentation: A Symbolic Dynamical Approach. Front Physiol 2017; 8:827. [PMID: 29184505 PMCID: PMC5694498 DOI: 10.3389/fphys.2017.00827] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/06/2017] [Indexed: 11/13/2022] Open
Abstract
Background: We recently introduced the concept of heart rate fragmentation along with a set of metrics for its quantification. The term was coined to refer to an increase in the percentage of changes in heart rate acceleration sign, a dynamical marker of a type of anomalous variability. The effort was motivated by the observation that fragmentation, which is consistent with the breakdown of the neuroautonomic-electrophysiologic control system of the sino-atrial node, could confound traditional short-term analysis of heart rate variability. Objective: The objectives of this study were to: (1) introduce a symbolic dynamical approach to the problem of quantifying heart rate fragmentation; (2) evaluate how the distribution of the different dynamical patterns (“words”) varied with the participants' age in a group of healthy subjects and patients with coronary artery disease (CAD); and (3) quantify the differences in the fragmentation patterns between the two sample populations. Methods: The symbolic dynamical method employed here was based on a ternary map of the increment NN interval time series and on the analysis of the relative frequency of symbolic sequences (words) with a pre-defined set of features. We analyzed annotated, open-access Holter databases of healthy subjects and patients with CAD, provided by the University of Rochester Telemetric and Holter ECG Warehouse (THEW). Results: The degree of fragmentation was significantly higher in older individuals than in their younger counterparts. However, the fragmentation patterns were different in the two sample populations. In healthy subjects, older age was significantly associated with a higher percentage of transitions from acceleration/deceleration to zero acceleration and vice versa (termed “soft” inflection points). In patients with CAD, older age was also significantly associated with higher percentages of frank reversals in heart rate acceleration (transitions from acceleration to deceleration and vice versa, termed “hard” inflection points). Compared to healthy subjects, patients with CAD had significantly higher percentages of soft and hard inflection points, an increased percentage of words with a high degree of fragmentation and a decreased percentage of words with a lower degree of fragmentation. Conclusion: The symbolic dynamical method employed here was useful to probe the newly recognized property of heart rate fragmentation. The findings from these cross-sectional studies confirm that CAD and older age are associated with higher levels of heart rate fragmentation. Furthermore, fragmentation with healthy aging appears to be phenotypically different from fragmentation in the context of CAD.
Collapse
Affiliation(s)
- Madalena D Costa
- Department of Medicine, Beth Israel Deaconess Medical Center, Margret and H. A. Rey Institute for Nonlinear Dynamics in Medicine, Harvard Medical School, Boston, MA, United States
| | - Roger B Davis
- Division of General Medicine and Primary Care, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ary L Goldberger
- Department of Medicine, Beth Israel Deaconess Medical Center, Margret and H. A. Rey Institute for Nonlinear Dynamics in Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
63
|
Affiliation(s)
- David Silbersweig
- Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
64
|
Costa MD, Davis RB, Goldberger AL. Heart Rate Fragmentation: A New Approach to the Analysis of Cardiac Interbeat Interval Dynamics. Front Physiol 2017; 8:255. [PMID: 28536533 PMCID: PMC5422439 DOI: 10.3389/fphys.2017.00255] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/10/2017] [Indexed: 02/02/2023] Open
Abstract
Background: Short-term heart rate variability (HRV) is most commonly attributed to physiologic vagal tone modulation. However, with aging and cardiovascular disease, the emergence of high short-term HRV, consistent with the breakdown of the neuroautonomic-electrophysiologic control system, may confound traditional HRV analysis. An apparent dynamical signature of such anomalous short-term HRV is frequent changes in heart rate acceleration sign, defined here as heart rate fragmentation. Objective: The aims were to: (1) introduce a set of metrics designed to probe the degree of sinus rhythm fragmentation; (2) test the hypothesis that the degree of fragmentation of heartbeat time series increases with the participants' age in a group of healthy subjects; (3) test the hypothesis that the heartbeat time series from patients with advanced coronary artery disease (CAD) are more fragmented than those from healthy subjects; and (4) compare the performance of the new fragmentation metrics with standard time and frequency domain measures of short-term HRV. Methods: We analyzed annotated, open-access Holter recordings (University of Rochester Holter Warehouse) from healthy subjects and patients with CAD using these newly introduced metrics of heart rate fragmentation, as well as standard time and frequency domain indices of short-term HRV, detrended fluctuation analysis and sample entropy. Results: The degree of fragmentation of cardiac interbeat interval time series increased significantly as a function of age in the healthy population as well as in patients with CAD. Fragmentation was higher for the patients with CAD than the healthy subjects. Heart rate fragmentation metrics outperformed traditional short-term HRV indices, as well as two widely used nonlinear measures, sample entropy and detrended fluctuation analysis short-term exponent, in distinguishing healthy subjects and patients with CAD. The same level of discrimination was obtained from the analysis of normal-to-normal sinus (NN) and cardiac interbeat interval (RR) time series. Conclusion: The fragmentation framework and accompanying metrics introduced here constitute a new way of assessing short-term HRV under free-running conditions, one which appears to overcome salient limitations of traditional HRV analysis. Fragmentation of sinus rhythm cadence may provide new dynamical biomarkers for probing the integrity of the neuroautonomic-electrophysiologic network controlling the heartbeat in health and disease.
Collapse
Affiliation(s)
- Madalena D Costa
- Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Roger B Davis
- Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Ary L Goldberger
- Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
65
|
Mitnitski AB, Rutenberg AD, Farrell S, Rockwood K. Aging, frailty and complex networks. Biogerontology 2017; 18:433-446. [PMID: 28255823 DOI: 10.1007/s10522-017-9684-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/21/2017] [Indexed: 12/21/2022]
Abstract
When people age their mortality rate increases exponentially, following Gompertz's law. Even so, individuals do not die from old age. Instead, they accumulate age-related illnesses and conditions and so become increasingly vulnerable to death from various external and internal stressors. As a measure of such vulnerability, frailty can be quantified using the frailty index (FI). Larger values of the FI are strongly associated with mortality and other adverse health outcomes. This association, and the insensitivity of the FI to the particular health variables that are included in its construction, makes it a powerful, convenient, and increasingly popular integrative health measure. Still, little is known about why the FI works so well. Our group has recently developed a theoretical network model of health deficits to better understand how changes in health are captured by the FI. In our model, health-related variables are represented by the nodes of a complex network. The network has a scale-free shape or "topology": a few nodes have many connections with other nodes, whereas most nodes have few connections. These nodes can be in two states, either damaged or undamaged. Transitions between damaged and non-damaged states are governed by the stochastic environment of individual nodes. Changes in the degree of damage of connected nodes change the local environment and make further damage more likely. Our model shows how age-dependent acceleration of the FI and of mortality emerges, even without specifying an age-damage relationship or any other time-dependent parameter. We have also used our model to assess how informative individual deficits are with respect to mortality. We find that the information is larger for nodes that are well connected than for nodes that are not. The model supports the idea that aging occurs as an emergent phenomenon, and not as a result of age-specific programming. Instead, aging reflects how damage propagates through a complex network of interconnected elements.
Collapse
Affiliation(s)
- A B Mitnitski
- Department of Medicine, Dalhousie University, Halifax, Canada.
- Geriatric Medicine Research Unit, Halifax, Canada.
| | - A D Rutenberg
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Canada
| | - S Farrell
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Canada
| | - K Rockwood
- Department of Medicine, Dalhousie University, Halifax, Canada
- Geriatric Medicine Research Unit, Halifax, Canada
| |
Collapse
|
66
|
Simple and complex retinal dystrophies are associated with profoundly different disease networks. Sci Rep 2017; 7:41835. [PMID: 28139756 PMCID: PMC5282568 DOI: 10.1038/srep41835] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/28/2016] [Indexed: 12/20/2022] Open
Abstract
Retinopathies are a group of monogenetic or complex retinal diseases associated with high unmet medical need. Monogenic disorders are caused by rare genetic variation and usually arise early in life. Other diseases, such as age-related macular degeneration (AMD), develop late in life and are considered to be of complex origin as they develop from a combination of genetic, ageing, environmental and lifestyle risk factors. Here, we contrast the underlying disease networks and pathological mechanisms of monogenic as opposed to complex retinopathies, using AMD as an example of the latter. We show that, surprisingly, genes associated with the different forms of retinopathies in general do not overlap despite their overlapping retinal phenotypes. Further, AMD risk genes participate in multiple networks with interaction partners that link to different ubiquitous pathways affecting general tissue integrity and homeostasis. Thus AMD most likely represents an endophenotype with differing underlying pathogenesis in different subjects. Localising these pathomechanisms and processes within and across different retinal anatomical compartments provides a novel representation of AMD that may be extended to complex disease in general. This approach may generate improved treatment options that target multiple processes with the aim of restoring tissue homeostasis and maintaining vision.
Collapse
|
67
|
Ferrari R, Lovering RC, Hardy J, Lewis PA, Manzoni C. Weighted Protein Interaction Network Analysis of Frontotemporal Dementia. J Proteome Res 2017; 16:999-1013. [PMID: 28004582 PMCID: PMC6152613 DOI: 10.1021/acs.jproteome.6b00934] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
The genetic analysis
of complex disorders has undoubtedly led to
the identification of a wealth of associations between genes and specific
traits. However, moving from genetics to biochemistry one gene at
a time has, to date, rather proved inefficient and under-powered to
comprehensively explain the molecular basis of phenotypes. Here we
present a novel approach, weighted protein–protein interaction
network analysis (W-PPI-NA), to highlight key functional players within
relevant biological processes associated with a given trait. This
is exemplified in the current study by applying W-PPI-NA to frontotemporal
dementia (FTD): We first built the state of the art FTD protein network
(FTD-PN) and then analyzed both its topological and functional features.
The FTD-PN resulted from the sum of the individual interactomes built
around FTD-spectrum genes, leading to a total of 4198 nodes. Twenty
nine of 4198 nodes, called inter-interactome hubs (IIHs), represented
those interactors able to bridge over 60% of the individual interactomes.
Functional annotation analysis not only reiterated and reinforced
previous findings from single genes and gene-coexpression analyses
but also indicated a number of novel potential disease related mechanisms,
including DNA damage response, gene expression
regulation, and cell waste disposal and
potential biomarkers or therapeutic targets including EP300. These
processes and targets likely represent the functional core impacted
in FTD, reflecting the underlying genetic architecture contributing
to disease. The approach presented in this study can be applied to
other complex traits for which risk-causative genes are known as it
provides a promising tool for setting the foundations for collating
genomics and wet laboratory data in a bidirectional manner. This is
and will be critical to accelerate molecular target prioritization
and drug discovery.
Collapse
Affiliation(s)
- Raffaele Ferrari
- Department of Molecular Neuroscience, UCL Institute of Neurology , Russell Square House, 9-12 Russell Square House, London WC1B 5EH, United Kingdom
| | - Ruth C Lovering
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London , London WC1E 6JF, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology , Russell Square House, 9-12 Russell Square House, London WC1B 5EH, United Kingdom
| | - Patrick A Lewis
- Department of Molecular Neuroscience, UCL Institute of Neurology , Russell Square House, 9-12 Russell Square House, London WC1B 5EH, United Kingdom.,School of Pharmacy, University of Reading , Whiteknights, Reading RG6 6AP, United Kingdom
| | - Claudia Manzoni
- Department of Molecular Neuroscience, UCL Institute of Neurology , Russell Square House, 9-12 Russell Square House, London WC1B 5EH, United Kingdom.,School of Pharmacy, University of Reading , Whiteknights, Reading RG6 6AP, United Kingdom
| |
Collapse
|
68
|
Udrescu L, Sbârcea L, Topîrceanu A, Iovanovici A, Kurunczi L, Bogdan P, Udrescu M. Clustering drug-drug interaction networks with energy model layouts: community analysis and drug repurposing. Sci Rep 2016; 6:32745. [PMID: 27599720 PMCID: PMC5013446 DOI: 10.1038/srep32745] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/12/2016] [Indexed: 11/15/2022] Open
Abstract
Analyzing drug-drug interactions may unravel previously unknown drug action patterns, leading to the development of new drug discovery tools. We present a new approach to analyzing drug-drug interaction networks, based on clustering and topological community detection techniques that are specific to complex network science. Our methodology uncovers functional drug categories along with the intricate relationships between them. Using modularity-based and energy-model layout community detection algorithms, we link the network clusters to 9 relevant pharmacological properties. Out of the 1141 drugs from the DrugBank 4.1 database, our extensive literature survey and cross-checking with other databases such as Drugs.com, RxList, and DrugBank 4.3 confirm the predicted properties for 85% of the drugs. As such, we argue that network analysis offers a high-level grasp on a wide area of pharmacological aspects, indicating possible unaccounted interactions and missing pharmacological properties that can lead to drug repositioning for the 15% drugs which seem to be inconsistent with the predicted property. Also, by using network centralities, we can rank drugs according to their interaction potential for both simple and complex multi-pathology therapies. Moreover, our clustering approach can be extended for applications such as analyzing drug-target interactions or phenotyping patients in personalized medicine applications.
Collapse
Affiliation(s)
- Lucreţia Udrescu
- “Victor Babeş” University of Medicine and Pharmacy Timişoara, Faculty of Pharmacy, Timişoara, 300041, Romania
| | - Laura Sbârcea
- “Victor Babeş” University of Medicine and Pharmacy Timişoara, Faculty of Pharmacy, Timişoara, 300041, Romania
| | - Alexandru Topîrceanu
- University Politehnica of Timişoara, Department of Computer and Information Technology, Timişoara, 300223, Romania
| | - Alexandru Iovanovici
- University Politehnica of Timişoara, Department of Computer and Information Technology, Timişoara, 300223, Romania
| | - Ludovic Kurunczi
- Institute of Chemistry Timişoara of the Romanian Academy, Timişoara, 300223, Romania
| | - Paul Bogdan
- University of Southern California, Ming Hsieh Department of Electrical Engineering, Los Angeles, CA 90089-2563, USA
| | - Mihai Udrescu
- University Politehnica of Timişoara, Department of Computer and Information Technology, Timişoara, 300223, Romania
| |
Collapse
|