51
|
Mead TJ, McCulloch DR, Ho JC, Du Y, Adams SM, Birk DE, Apte SS. The metalloproteinase-proteoglycans ADAMTS7 and ADAMTS12 provide an innate, tendon-specific protective mechanism against heterotopic ossification. JCI Insight 2018; 3:92941. [PMID: 29618652 DOI: 10.1172/jci.insight.92941] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/28/2018] [Indexed: 12/22/2022] Open
Abstract
Heterotopic ossification (HO) is a significant clinical problem with incompletely resolved mechanisms. Here, the secreted metalloproteinases ADAMTS7 and ADAMTS12 are shown to comprise a unique proteoglycan class that protects against a tendency toward HO in mouse hindlimb tendons, menisci, and ligaments. Adamts7 and Adamts12 mRNAs were sparsely expressed in murine forelimbs but strongly coexpressed in hindlimb tendons, skeletal muscle, ligaments, and meniscal fibrocartilage. Adamts7-/- Adamts12-/- mice, but not corresponding single-gene mutants, which demonstrated compensatory upregulation of the intact homolog mRNA, developed progressive HO in these tissues after 4 months of age. Adamts7-/- Adamts12-/- tendons had abnormal collagen fibrils, accompanied by reduced levels of the small leucine-rich proteoglycans (SLRPs) biglycan, fibromodulin, and decorin, which regulate collagen fibrillogenesis. Bgn-/0 Fmod-/- mice are known to have a strikingly similar hindlimb HO to that of Adamts7-/- Adamts12-/- mice, implicating fibromodulin and biglycan reduction as a likely mechanism underlying HO in Adamts7-/- Adamts12-/- mice. Interestingly, degenerated human biceps tendons had reduced ADAMTS7 mRNA compared with healthy biceps tendons, which expressed both ADAMTS7 and ADAMTS12. These results suggest that ADAMTS7 and ADAMTS12 drive an innate pathway protective against hindlimb HO in mice and may be essential for human tendon health.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Daniel R McCulloch
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Jason C Ho
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA.,Department of Orthopaedic Surgery and the Orthopaedic and Rheumatology Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yaoyao Du
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Sheila M Adams
- Departments of Molecular Pharmacology and Physiology and Orthopaedics and Sports Medicine, University of South Florida, Morsani College of Medicine, Tampa, Florida, USA
| | - David E Birk
- Departments of Molecular Pharmacology and Physiology and Orthopaedics and Sports Medicine, University of South Florida, Morsani College of Medicine, Tampa, Florida, USA
| | - Suneel S Apte
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| |
Collapse
|
52
|
Kowara M, Cudnoch-Jedrzejewska A, Opolski G, Wlodarski P. MicroRNA regulation of extracellular matrix components in the process of atherosclerotic plaque destabilization. Clin Exp Pharmacol Physiol 2018; 44:711-718. [PMID: 28440887 DOI: 10.1111/1440-1681.12772] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/23/2017] [Accepted: 04/19/2017] [Indexed: 12/13/2022]
Abstract
The process of atherosclerotic plaque destabilization, leading to myocardial infarction, is still not fully understood. The pathway - composed of structural and regulatory proteins of the extracellular matrix (ECM) such as collagen, elastin, small leucine-rich proteoglycans, metalloproteinases, cathepsins and serine proteases - is one potential way of atherosclerotic plaque destabilization. The expression of these proteins is controlled by different microRNA molecules. The goal of this paper is to summarize the current investigations and knowledge about ECM in the process of atherosclerotic plaque destabilization, giving special attention to epigenetic expression regulation by microRNA.
Collapse
Affiliation(s)
- Michal Kowara
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.,First Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jedrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz Opolski
- First Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Pawel Wlodarski
- Department of Histology and Embryology, Center for Biostructure Research, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
53
|
Metalloproteinases in atherosclerosis. Eur J Pharmacol 2017; 816:93-106. [DOI: 10.1016/j.ejphar.2017.09.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/31/2017] [Accepted: 09/08/2017] [Indexed: 11/20/2022]
|
54
|
Chan K, Pu X, Sandesara P, Poston RN, Simpson IA, Quyyumi AA, Ye S, Patel RS. Genetic Variation at the ADAMTS7 Locus is Associated With Reduced Severity of Coronary Artery Disease. J Am Heart Assoc 2017; 6:JAHA.117.006928. [PMID: 29089340 PMCID: PMC5721775 DOI: 10.1161/jaha.117.006928] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Genome-wide association studies identified ADAMTS7 as a risk locus for coronary artery disease (CAD). Functional studies suggest that ADAMTS7 may promote cellular processes in atherosclerosis. We sought to examine the association between genetic variation at ADAMTS7 and measures of atherosclerosis using histological, angiographic, and clinical outcomes data. METHODS AND RESULTS The lead CAD-associated single-nucleotide polymorphism rs3825807 at the ADAMTS7 locus was genotyped. The G allele (reduced ADAMTS7 function) was associated with a smaller fibrous cap (P=0.017) and a smaller percentage area of α-actin (smooth muscle cell marker) in the intima (P=0.017), but was not associated with calcification or plaque thickness, following ex vivo immunohistochemistry analysis of human coronary plaques (n=50; mean age 72.2±11.3). In two independent cohorts (Southampton Atherosclerosis Study [n=1359; mean age 62.5±10.3; 70.1% men] and the Emory Cardiovascular Biobank [EmCAB; n=2684; mean age 63.8±11.3; 68.7% men]), the G allele was associated with 16% to 19% lower odds of obstructive CAD (Southampton Atherosclerosis Study: odds ratio, 0.81; 95% confidence interval, 0.67-0.98; EmCAB: odds ratio, 0.84; 95% confidence interval, 0.75-0.95) with similar effects for multivessel, left anterior descending, and proximal CAD. Furthermore, each copy of the G allele was associated with lower angiographic severity Gensini score (Southampton Atherosclerosis Study, P=0.026; EmCAB, P<0.001), lower Sullivan Extent score (Southampton Atherosclerosis Study, P=0.029; EmCAB, P<0.001), and a 23% lower risk of incident revascularization procedures (EmCAB: hazard ratio, 0.76; 95% confidence interval, 0.59-0.98). There were no associations with all-cause mortality or incident myocardial infarction. CONCLUSIONS Genetic variation at the ADAMTS7 locus is associated with several complementary CAD phenotypes, supporting the emerging role of ADAMTS7 in atherosclerosis and may represent a potential drug target.
Collapse
Affiliation(s)
- Kenneth Chan
- Institute of Cardiovascular Sciences, University College London, London, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Xiangyuan Pu
- William Harvey Research Institute, Queen Mary University of London, United Kingdom
| | - Pratik Sandesara
- Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Robin N Poston
- William Harvey Research Institute, Queen Mary University of London, United Kingdom
| | - Iain A Simpson
- Wessex Regional Cardiac Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Arshed A Quyyumi
- Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Shu Ye
- Department of Cardiovascular Sciences, University of Leicester, United Kingdom
- Shantou University Medical College, Shantou, China
| | - Riyaz S Patel
- Institute of Cardiovascular Sciences, University College London, London, United Kingdom
- Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| |
Collapse
|
55
|
Lacolley P, Regnault V, Segers P, Laurent S. Vascular Smooth Muscle Cells and Arterial Stiffening: Relevance in Development, Aging, and Disease. Physiol Rev 2017; 97:1555-1617. [DOI: 10.1152/physrev.00003.2017] [Citation(s) in RCA: 332] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/15/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
The cushioning function of large arteries encompasses distension during systole and recoil during diastole which transforms pulsatile flow into a steady flow in the microcirculation. Arterial stiffness, the inverse of distensibility, has been implicated in various etiologies of chronic common and monogenic cardiovascular diseases and is a major cause of morbidity and mortality globally. The first components that contribute to arterial stiffening are extracellular matrix (ECM) proteins that support the mechanical load, while the second important components are vascular smooth muscle cells (VSMCs), which not only regulate actomyosin interactions for contraction but mediate also mechanotransduction in cell-ECM homeostasis. Eventually, VSMC plasticity and signaling in both conductance and resistance arteries are highly relevant to the physiology of normal and early vascular aging. This review summarizes current concepts of central pressure and tensile pulsatile circumferential stress as key mechanical determinants of arterial wall remodeling, cell-ECM interactions depending mainly on the architecture of cytoskeletal proteins and focal adhesion, the large/small arteries cross-talk that gives rise to target organ damage, and inflammatory pathways leading to calcification or atherosclerosis. We further speculate on the contribution of cellular stiffness along the arterial tree to vascular wall stiffness. In addition, this review provides the latest advances in the identification of gene variants affecting arterial stiffening. Now that important hemodynamic and molecular mechanisms of arterial stiffness have been elucidated, and the complex interplay between ECM, cells, and sensors identified, further research should study their potential to halt or to reverse the development of arterial stiffness.
Collapse
Affiliation(s)
- Patrick Lacolley
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Véronique Regnault
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Patrick Segers
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Stéphane Laurent
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| |
Collapse
|
56
|
Orentas RJ, Sindiri S, Duris C, Wen X, He J, Wei JS, Jarzembowski J, Khan J. Paired Expression Analysis of Tumor Cell Surface Antigens. Front Oncol 2017; 7:173. [PMID: 28871274 PMCID: PMC5566986 DOI: 10.3389/fonc.2017.00173] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/31/2017] [Indexed: 01/15/2023] Open
Abstract
Adoptive immunotherapy with antibody-based therapy or with T cells transduced to express chimeric antigen receptors (CARs) is useful to the extent that the cell surface membrane protein being targeted is not expressed on normal tissues. The most successful CAR-based (anti-CD19) or antibody-based therapy (anti-CD20) in hematologic malignancies has the side effect of eliminating the normal B cell compartment. Targeting solid tumors may not provide a similar expendable marker. Beyond antibody to Her2/NEU and EGFR, very few antibody-based and no CAR-based therapies have seen broad clinical application for solid tumors. To expand the way in which the surfaceome of solid tumors can be analyzed, we created an algorithm that defines the pairwise relative overexpression of surface antigens. This enables the development of specific immunotherapies that require the expression of two discrete antigens on the surface of the tumor target. This dyad analysis was facilitated by employing the Hotelling’s T-squared test (Hotelling–Lawley multivariate analysis of variance) for two independent variables in comparison to a third constant entity (i.e., gene expression levels in normal tissues). We also present a unique consensus scoring mechanism for identifying transcripts that encode cell surface proteins. The unique application of our bioinformatics processing pipeline and statistical tools allowed us to compare the expression of two membrane protein targets as a pair, and to propose a new strategy based on implementing immunotherapies that require both antigens to be expressed on the tumor cell surface to trigger therapeutic effector mechanisms. Specifically, we found that, for MYCN amplified neuroblastoma, pairwise expression of ACVR2B or anaplastic lymphoma kinase (ALK) with GFRA3, GFRA2, Cadherin 24, or with one another provided the strongest hits. For MYCN, non-amplified stage 4 neuroblastoma, neurotrophic tyrosine kinase 1, or ALK paired with GFRA2, GFRA3, SSK1, GPR173, or with one another provided the most promising paired-hits. We propose that targeting these markers together would increase the specificity and thereby the safety of CAR-based therapy for neuroblastoma.
Collapse
Affiliation(s)
- Rimas J Orentas
- Lentigen Technology, Inc., a Miltenyi Biotec Company, Gaithersburg, MD, United States
| | - Sivasish Sindiri
- Genetics Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Christine Duris
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Xinyu Wen
- Genetics Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Jianbin He
- Genetics Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Jun S Wei
- Genetics Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Jason Jarzembowski
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Javed Khan
- Genetics Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
57
|
Abstract
Several large-scale genome-wide association studies have identified single-nucleotide polymorphisms in the genomic region of A Disintegrin And Metalloproteinase with ThromboSpondin type 1 repeats (ADAMTS)-7 and associations to coronary artery disease. Experimental studies have provided evidence for a functional role of ADAMTS-7 in both injury-induced vascular neointima formation and development of atherosclerotic lesions. However, whether ADAMTS-7 is associated with a specific plaque phenotype in humans has not been investigated. Carotid plaques (n = 206) from patients with and without cerebrovascular symptoms were analyzed for expression of ADAMTS-7 by immunohistochemistry and correlated to components associated with plaque vulnerability. Plaques from symptomatic patients showed increased levels of ADAMTS-7 compared with lesions from asymptomatic patients. High levels of ADAMTS-7 correlated with high levels of CD68-staining and lipid content, but with low smooth muscle cell and collagen content, which together are characteristics of a vulnerable plaque phenotype. ADAMTS-7 levels above median were associated with increased risk for postoperative cardiovascular events. Our data show that ADAMTS-7 is associated with a vulnerable plaque phenotype in human carotid lesions. These data support previous observations of a potential proatherogenic role of ADAMTS-7.
Collapse
|
58
|
Saleheen D, Zhao W, Young R, Nelson CP, Ho W, Ferguson JF, Rasheed A, Ou K, Nurnberg ST, Bauer RC, Goel A, Do R, Stewart AF, Hartiala J, Zhang W, Thorleifsson G, Strawbridge RJ, Sinisalo J, Kanoni S, Sedaghat S, Marouli E, Kristiansson K, Zhao JH, Scott R, Gauguier D, Shah SH, Smith AV, van Zuydam N, Cox AJ, Willenborg C, Kessler T, Zeng L, Province MA, Ganna A, Lind L, Pedersen NL, White CC, Joensuu A, Kleber ME, Hall AS, März W, Salomaa V, O’Donnell C, Ingelsson E, Feitosa MF, Erdmann J, Bowden DW, Palmer CN, Gudnason V, De Faire U, Zalloua P, Wareham N, Thompson JR, Kuulasmaa K, Dedoussis G, Perola M, Dehghan A, Chambers JC, Kooner J, Allayee H, Deloukas P, McPherson R, Stefansson K, Schunkert H, Kathiresan S, Farrall M, Frossard PM, Rader DJ, Samani NJ, Reilly MP. Loss of Cardioprotective Effects at the ADAMTS7 Locus as a Result of Gene-Smoking Interactions. Circulation 2017; 135:2336-2353. [PMID: 28461624 PMCID: PMC5612779 DOI: 10.1161/circulationaha.116.022069] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 03/21/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND Common diseases such as coronary heart disease (CHD) are complex in etiology. The interaction of genetic susceptibility with lifestyle factors may play a prominent role. However, gene-lifestyle interactions for CHD have been difficult to identify. Here, we investigate interaction of smoking behavior, a potent lifestyle factor, with genotypes that have been shown to associate with CHD risk. METHODS We analyzed data on 60 919 CHD cases and 80 243 controls from 29 studies for gene-smoking interactions for genetic variants at 45 loci previously reported to be associated with CHD risk. We also studied 5 loci associated with smoking behavior. Study-specific gene-smoking interaction effects were calculated and pooled using fixed-effects meta-analyses. Interaction analyses were declared to be significant at a P value of <1.0×10-3 (Bonferroni correction for 50 tests). RESULTS We identified novel gene-smoking interaction for a variant upstream of the ADAMTS7 gene. Every T allele of rs7178051 was associated with lower CHD risk by 12% in never-smokers (P=1.3×10-16) in comparison with 5% in ever-smokers (P=2.5×10-4), translating to a 60% loss of CHD protection conferred by this allelic variation in people who smoked tobacco (interaction P value=8.7×10-5). The protective T allele at rs7178051 was also associated with reduced ADAMTS7 expression in human aortic endothelial cells and lymphoblastoid cell lines. Exposure of human coronary artery smooth muscle cells to cigarette smoke extract led to induction of ADAMTS7. CONCLUSIONS: Allelic variation at rs7178051 that associates with reduced ADAMTS7 expression confers stronger CHD protection in never-smokers than in ever-smokers. Increased vascular ADAMTS7 expression may contribute to the loss of CHD protection in smokers.
Collapse
Affiliation(s)
- Danish Saleheen
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Wei Zhao
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA
| | - Robin Young
- Department of Public Health and Primary Care, University of Cambridge, United Kingdom
| | - Christopher P. Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - WeangKee Ho
- Department of Public Health and Primary Care, University of Cambridge, United Kingdom
| | - Jane F. Ferguson
- Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN
| | - Asif Rasheed
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Kristy Ou
- Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN
| | - Sylvia T. Nurnberg
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Robert C. Bauer
- Cardiology Division, Department of Medicine and the Irving Institute for Clinical and Translational Research, Columbia University Medical Center, New York, NY
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine & Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ron Do
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexandre F.R. Stewart
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Canada
| | - Jaana Hartiala
- Institute for Genetic Medicine and Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Department of Cardiology, Ealing Hospital NHS Trust, Middlesex, United Kingdom
| | - Gudmar Thorleifsson
- deCODE Genetics, Sturlugata 8, IS-101 Reykjavik, Iceland
- University of Iceland, School of Medicine, Reykjavik, Iceland
| | - Rona J Strawbridge
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Stavroula Kanoni
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sanaz Sedaghat
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Eirini Marouli
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Dietetics-Nutrition, Harokopio University, 70 El. VenizelouStr, Athens, Greece
| | | | - Jing Hua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Robert Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | | | - Svati H. Shah
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Natalie van Zuydam
- Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Amanda J. Cox
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC
| | - Christina Willenborg
- Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany
- DZHK (German Research Center for Cardiovascular Research) partner site Hamburg–Lübeck–Kiel, Lübeck, Germany
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Technische Universität München, München, Germany
- Klinikum rechts der Isar, München, Germany
| | - Lingyao Zeng
- Deutsches Herzzentrum München, Technische Universität München, München, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, München, Germany
| | - Michael A. Province
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Andrea Ganna
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Nancy L. Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Charles C. White
- Department of Biostatistics Boston University School of Public Health Framingham Heart Study, Boston, MA
| | - Anni Joensuu
- National Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki, Institute for Molecular Medicine, Finland (FIMM)
| | - Marcus Edi Kleber
- Department of Medicine, Mannheim Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Alistair S. Hall
- Leeds Institute of Genetics, Health and Therapeutics, University of Leeds, Leeds, United Kingdom
| | - Winfried März
- Synlab Academy, Synlab Services GmbH, Mannheim, Germany and Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Christopher O’Donnell
- National Heart, Lung, and Blood Institute and the Framingham Heart Study, National Institutes of Health, Bethesda, MD
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
| | - Mary F. Feitosa
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Jeanette Erdmann
- Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany
- DZHK (German Research Center for Cardiovascular Research) partner site Hamburg–Lübeck–Kiel, Lübeck, Germany
| | - Donald W. Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC
| | - Colin N.A. Palmer
- Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ulf De Faire
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Pierre Zalloua
- Lebanese American University, School of Medicine, Beirut, Lebanon
| | - Nicholas Wareham
- INSERM, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
| | - John R. Thompson
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Kari Kuulasmaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - George Dedoussis
- Department of Dietetics-Nutrition, Harokopio University, 70 El. VenizelouStr, Athens, Greece
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki, Institute for Molecular Medicine, Finland (FIMM)
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John C. Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Department of Cardiology, Ealing Hospital NHS Trust, Middlesex, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Jaspal Kooner
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
- Cardiovascular Science, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Hooman Allayee
- Institute for Genetic Medicine and Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Panos Deloukas
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ruth McPherson
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Canada
| | - Kari Stefansson
- deCODE Genetics, Sturlugata 8, IS-101 Reykjavik, Iceland
- University of Iceland, School of Medicine, Reykjavik, Iceland
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, München, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, München, Germany
| | - Sekar Kathiresan
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine & Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - EPIC-CVD
- Department of Public Health and Primary Care, University of Cambridge, United Kingdom
| | | | - Daniel J. Rader
- Department of Genetics, University of Pennsylvania, Philadelphia, PA
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - PROMIS
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | | | - Muredach P. Reilly
- Cardiology Division, Department of Medicine and the Irving Institute for Clinical and Translational Research, Columbia University Medical Center, New York, NY
| |
Collapse
|
59
|
Khera AV, Kathiresan S. Genetics of coronary artery disease: discovery, biology and clinical translation. Nat Rev Genet 2017; 18:331-344. [PMID: 28286336 PMCID: PMC5935119 DOI: 10.1038/nrg.2016.160] [Citation(s) in RCA: 403] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Coronary artery disease is the leading global cause of mortality. Long recognized to be heritable, recent advances have started to unravel the genetic architecture of the disease. Common variant association studies have linked approximately 60 genetic loci to coronary risk. Large-scale gene sequencing efforts and functional studies have facilitated a better understanding of causal risk factors, elucidated underlying biology and informed the development of new therapeutics. Moving forwards, genetic testing could enable precision medicine approaches by identifying subgroups of patients at increased risk of coronary artery disease or those with a specific driving pathophysiology in whom a therapeutic or preventive approach would be most useful.
Collapse
Affiliation(s)
- Amit V Khera
- Division of Cardiology, Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital; Cardiovascular Disease Initiative, Broad Institute of Harvard and Massachusetts Institute of Technology, 185 Cambridge Street, CPZN 5.252, Boston, Massachusetts 02114, USA
| | - Sekar Kathiresan
- Division of Cardiology, Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital; Cardiovascular Disease Initiative, Broad Institute of Harvard and Massachusetts Institute of Technology, 185 Cambridge Street, CPZN 5.252, Boston, Massachusetts 02114, USA
| |
Collapse
|
60
|
Warren HR, Evangelou E, Cabrera CP, Gao H, Ren M, Mifsud B, Ntalla I, Surendran P, Liu C, Cook JP, Kraja AT, Drenos F, Loh M, Verweij N, Marten J, Karaman I, Segura Lepe MP, O’Reilly PF, Knight J, Snieder H, Kato N, He J, Tai ES, Said MA, Porteous D, Alver M, Poulter N, Farrall M, Gansevoort RT, Padmanabhan S, Mägi R, Stanton A, Connell J, Bakker SJL, Metspalu A, Shields DC, Thom S, Brown M, Sever P, Esko T, Hayward C, van der Harst P, Saleheen D, Chowdhury R, Chambers JC, Chasman DI, Chakravarti A, Newton-Cheh C, Lindgren CM, Levy D, Kooner JS, Keavney B, Tomaszewski M, Samani NJ, Howson JMM, Tobin MD, Munroe PB, Ehret GB, Wain LV, Barnes MR, Tzoulaki I, Caulfield MJ, Elliott P. Genome-wide association analysis identifies novel blood pressure loci and offers biological insights into cardiovascular risk. Nat Genet 2017; 49:403-415. [PMID: 28135244 PMCID: PMC5972004 DOI: 10.1038/ng.3768] [Citation(s) in RCA: 394] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/14/2016] [Indexed: 11/21/2022]
Abstract
Elevated blood pressure is the leading heritable risk factor for cardiovascular disease worldwide. We report genetic association of blood pressure (systolic, diastolic, pulse pressure) among UK Biobank participants of European ancestry with independent replication in other cohorts, and robust validation of 107 independent loci. We also identify new independent variants at 11 previously reported blood pressure loci. In combination with results from a range of in silico functional analyses and wet bench experiments, our findings highlight new biological pathways for blood pressure regulation enriched for genes expressed in vascular tissues and identify potential therapeutic targets for hypertension. Results from genetic risk score models raise the possibility of a precision medicine approach through early lifestyle intervention to offset the impact of blood pressure-raising genetic variants on future cardiovascular disease risk.
Collapse
Affiliation(s)
| | - Helen R Warren
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Claudia P Cabrera
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - He Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Meixia Ren
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - Borbala Mifsud
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Praveen Surendran
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Chunyu Liu
- Population Sciences Branch, National Heart Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Boston University School of Public Health, Boston, MA, USA
- National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis MO, USA
| | - Fotios Drenos
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, UK
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, Rayne Building, University College London, London, WC1E 6JF, UK
| | - Marie Loh
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, USA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan Marten
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Ibrahim Karaman
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Marcelo P Segura Lepe
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- Bayer Pharma AG, Berlin, Germany
| | - Paul F O’Reilly
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Joanne Knight
- Data Science Institute, Lancester University, Lancaster, UK
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - M Abdullah Said
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - David Porteous
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Maris Alver
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Neil Poulter
- Imperial Clinical Trials Unit, School of Public Health, Imperial College London, London, UK
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ron T Gansevoort
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Alice Stanton
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - John Connell
- Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Stephan J L Bakker
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Denis C Shields
- School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Simon Thom
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Morris Brown
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - Peter Sever
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Tõnu Esko
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, USA
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Caroline Hayward
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Danish Saleheen
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, USA
- Centre for Non-Communicable Diseases, Karachi, Pakistan
- Department of Public Health and Primary Care, University of Cambridge, UK
| | - Rajiv Chowdhury
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - John C Chambers
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher Newton-Cheh
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, USA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Cecilia M Lindgren
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, USA
- Wellcome Trust Center for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- The Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7BN, UK
| | - Daniel Levy
- Population Sciences Branch, National Heart Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - Jaspal S Kooner
- Imperial College Healthcare NHS Trust, London, UK
- Department of Cardiology, Ealing Hospital NHS Trust, Southall, Middlesex, UK
- National Heart and Lung Institute, Cardiovascular Sciences, Hammersmith Campus, Imperial College London, London, UK
| | - Bernard Keavney
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Division of Medicine, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Division of Medicine, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Joanna M M Howson
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Martin D Tobin
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - Georg B Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Cardiology, Department of Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Louise V Wain
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Michael R Barnes
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Mark J Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Bayoglu B, Arslan C, Tel C, Ulutin T, Dirican A, Deser SB, Cengiz M. Genetic variants rs1994016 and rs3825807 in ADAMTS7 affect its mRNA expression in atherosclerotic occlusive peripheral arterial disease. J Clin Lab Anal 2017; 32. [PMID: 28205274 DOI: 10.1002/jcla.22174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 01/18/2017] [Indexed: 11/07/2022] Open
Abstract
AIM Peripheral artery disease (PAD) is a vascular disease affecting peripheral circulation. Recently, genome-wide association studies revealed a relationship between single nucleotide polymorphisms (SNPs) in ADAMTS7 (a disintegrin and metalloprotease with thrombospondin motif 7) and atherosclerosis. In this study, we aimed to determine ADAMTS7 expression in peripheral blood mononuclear cells (PBMCs) and the frequency of ADAMTS7 rs1994016 and rs3825807 polymorphisms in a sample of Turkish patients with PAD, and to evaluate the association of matrix metalloproteinase (MMP) levels with PAD development. METHODS In this case-control study, ADAMTS7mRNA and protein expression was determined using reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) and western blot, respectively, and rs1994016 and rs3825807 variants in ADAMTS7 were determined by real-time PCR in 115 PAD patients and 116 healthy controls. Plasma levels of nine MMPs were determined using a multiplex immunoassay system. RESULTS ADAMTS7mRNA levels were significantly higher in PAD patients than in controls (t=-2.75, P=.007). There was no significant difference in the frequencies of rs1994016 and rs3825807 between PAD patients and controls (P>.05). In PAD patients, ADAMTS7mRNA levels were significantly increased for the CC genotype of rs1994016 (t=-2.31, P=.026) and TT genotype of rs3825807 (t=-2.23, P=.032). Furthermore, plasma levels of MMP-1, MMP-3, MMP-7, MMP-10, MMP-12, and MMP-13 were significantly higher in PAD patients than in controls (P<.05). CONCLUSION This is the first report of the relationship between PAD and ADAMTS7 expression and the effects of the rs1994016 and rs3825807 variants on PAD development. ADAMTS7 may be associated with PAD development.
Collapse
Affiliation(s)
- Burcu Bayoglu
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Caner Arslan
- Department of Cardiovascular Surgery, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Cigdem Tel
- Department of Cardiovascular Surgery, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Turgut Ulutin
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Ahmet Dirican
- Department of Biostatistics and Medical Informatics, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Serkan Burc Deser
- Department of Cardiovascular Surgery, Medical Faculty, Ondokuz Mayis University, Samsun, Turkey
| | - Mujgan Cengiz
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
62
|
Yu J, Zhou B, Yu H, Han J, Cui M, Zhang F, Wang G, Guo L, Gao W. Association between plasma ADAMTS-7 levels and severity of disease in patients with stable obstructive coronary artery disease. Medicine (Baltimore) 2016; 95:e5523. [PMID: 27902619 PMCID: PMC5134802 DOI: 10.1097/md.0000000000005523] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The metalloproteinase family of a disintegrin and metalloproteinase with thrombospondin motifs-7 (ADAMTS-7) was reported to be a novel locus associated with human coronary artery disease. This study aimed to investigate plasma ADAMTS-7 levels in stable obstructive CAD patients and elucidate the relationship between plasma ADAMTS-7 levels and the severity of CAD assessed by the Syntax score.This was a single center cross-sectional study performed in 182 CAD patients. ELISA was used to measure plasma ADAMTS-7 levels. All patients were divided into subgroup according to the ADAMTS-7 median in this cohort: high group with ADAMTS-7 ≥0.99 ng/mL and low group with ADAMTS-7 <0.99 ng/mL. Furthermore, all patients were divided into tertiles according to their Syntax scores (low group: Syntax score ≤10.0; moderate group: 10.0 <Syntax score ≤18.0; high group: Syntax score >18.0). We followed up the participants continuously until the first major adverse cardiovascular event (MACE) for a mean time of 22.0 months.Plasma ADAMTS-7 levels in the high Syntax score group were significantly higher compared with the low Syntax score group (3.29 [0.08-26.3] ng/mL vs 1.24 [0.15-8.78] ng/mL, P = 0.010). Plasma ADAMTS-7 levels were significantly positively correlated with the Syntax score tertiles (r = 0.157, P = 0.035). Logistic regression analysis indicated that the plasma ADAMTS-7 level was one of the independent predictors for the Syntax score tertiles (B = 1.118, 95% CI: 1.194-7.830, P = 0.020), together with HbA1c (B = 0.946, 95% CI: 1.248-5.312, P = 0.010), uric acid (B = -0.019, 95% CI: 0.974-0.988, P<0.001), and coronary artery calcium score (B = -0.001, 95% CI: 0.998-0.999, P < 0.001). Compared with the low ADAMTS-7 group, the high ADAMTS-7 group had significantly higher Syntax score (17.10±8.42 vs 14.96 ± 8.11, P = 0.047). Kaplan-Meier analysis showed patients in the high plasma ADAMTS-7 group tend to have a lower event-free survival rate than patients in the low plasma ADAMTS-7 group, unfortunately, no difference was detected (86.8% vs 88.0%, log rank = 0.314, P = 0.575).The plasma ADAMTS-7 level was positively correlated with the Syntax score significantly. The elevated plasma ADAMTS-7 level may be involved in the severity of disease in patients with stable coronary artery disease.
Collapse
|
63
|
Pereira A, Palma dos Reis R, Rodrigues R, Sousa AC, Gomes S, Borges S, Ornelas I, Freitas AI, Guerra G, Henriques E, Rodrigues M, Freitas S, Freitas C, Brehm A, Pereira D, Mendonça MI. Association of ADAMTS7 gene polymorphism with cardiovascular survival in coronary artery disease. Physiol Genomics 2016; 48:810-815. [DOI: 10.1152/physiolgenomics.00059.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 09/08/2016] [Indexed: 12/13/2022] Open
Abstract
Recent genetic studies have revealed an association between polymorphisms at the ADAMTS7 gene locus and coronary artery disease (CAD) risk. Functional studies have shown that a CAD-associated polymorphism (rs3825807) affects ADAMTS7 maturation and vascular smooth muscular cell (VSMC) migration. Here, we tested whether ADAMTS7 (A/G) SNP is associated with cardiovascular (CV) survival in patients with established CAD. A cohort of 1,128 patients with angiographic proven CAD, who were followed up prospectively for a mean follow-up period of 63 (range 6–182) mo, were genotyped for rs3825807 A/G. Survival statistics (Cox regression) compared heterozygous (AG) and wild-type (AA) with the reference homozygous GG. Kaplan-Meier (K-M) survival curves were performed according to ADAMTS7 genotypes for CV mortality. Results showed that 47.3% of patients were heterozygous (AG), 36.5% were homozygous for the wild-type allele (AA) and only 16.2% were homozygous for the GG genotype. During the follow-up period, 109 (9.7%) patients died, 77 (6.8%) of CV causes. Survival analysis showed that AA genotype was an independent risk factor for CV mortality compared with reference genotype GG (HR = 2.7, P = 0.025). At the end of follow-up, the estimated survival probability (K-M) was 89.8% for GG genotype, 82.2% for AG and 72.3% for AA genotype ( P = 0.039). Carriage of the mutant G allele of the ADAMTS7 gene was associated with improved CV survival in patients with documented CAD. The native overfunctional ADAMTS7 allele (A) may accelerate VSMC migration and lead to neointimal thickening, atherosclerosis progression and acute plaque events. ADAMTS7 gene should be further explored in CAD for risk prediction, mechanistic and therapeutic goals.
Collapse
Affiliation(s)
- A. Pereira
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - R. Palma dos Reis
- Faculty of Medical Sciences, New University of Lisbon, Lisbon, Portugal; and
| | - R. Rodrigues
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - A. C. Sousa
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - S. Gomes
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - S. Borges
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - I. Ornelas
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - A. I. Freitas
- Laboratory of Human Genetics, Madeira University, Campus da Penteada, Funchal, Madeira, Portugal
| | - G. Guerra
- Laboratory of Human Genetics, Madeira University, Campus da Penteada, Funchal, Madeira, Portugal
| | - E. Henriques
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - M. Rodrigues
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - S. Freitas
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - C. Freitas
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - A. Brehm
- Laboratory of Human Genetics, Madeira University, Campus da Penteada, Funchal, Madeira, Portugal
| | - D. Pereira
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| | - M. I. Mendonça
- Funchal Hospital Center, Research Unit and Cardiology Department, Funchal, Madeira, Portugal
| |
Collapse
|
64
|
Abstract
Genetic factors contribute importantly to the risk of coronary artery disease (CAD), and in the past decade, there has been major progress in this area. The tools applied include genome-wide association studies encompassing >200,000 individuals complemented by bioinformatic approaches, including 1000 Genomes imputation, expression quantitative trait locus analyses, and interrogation of Encyclopedia of DNA Elements, Roadmap, and other data sets. close to 60 common SNPs (minor allele frequency>0.05) associated with CAD risk and reaching genome-wide significance (P<5 × 10(-8)) have been identified. Furthermore, a total of 202 independent signals in 109 loci have achieved a false discovery rate (q<0.05) and together explain 28% of the estimated heritability of CAD. These data have been used successfully to create genetic risk scores that can improve risk prediction beyond conventional risk factors and identify those individuals who will benefit most from statin therapy. Such information also has important applications in clinical medicine and drug discovery by using a Mendelian randomization approach to interrogate the causal nature of many factors found to associate with CAD risk in epidemiological studies. In contrast to genome-wide association studies, whole-exome sequencing has provided valuable information directly relevant to genes with known roles in plasma lipoprotein metabolism but has, thus far, failed to identify other rare coding variants linked to CAD. Overall, recent studies have led to a broader understanding of the genetic architecture of CAD and demonstrate that it largely derives from the cumulative effect of multiple common risk alleles individually of small effect size rather than rare variants with large effects on CAD risk. Despite this success, there has been limited progress in understanding the function of the novel loci; the majority of which are in noncoding regions of the genome.
Collapse
Affiliation(s)
- Ruth McPherson
- From the Department of Medicine, Atherogenomics Laboratory, Division of Cardiology, Ruddy Canadian Cardiovascular Genetics Center, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.M.); and Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.T.-H.).
| | - Anne Tybjaerg-Hansen
- From the Department of Medicine, Atherogenomics Laboratory, Division of Cardiology, Ruddy Canadian Cardiovascular Genetics Center, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.M.); and Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.T.-H.)
| |
Collapse
|
65
|
Nurnberg ST, Zhang H, Hand NJ, Bauer RC, Saleheen D, Reilly MP, Rader DJ. From Loci to Biology: Functional Genomics of Genome-Wide Association for Coronary Disease. Circ Res 2016; 118:586-606. [PMID: 26892960 DOI: 10.1161/circresaha.115.306464] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genome-wide association studies have provided a rich collection of ≈ 58 coronary artery disease (CAD) loci that suggest the existence of previously unsuspected new biology relevant to atherosclerosis. However, these studies only identify genomic loci associated with CAD, and many questions remain even after a genomic locus is definitively implicated, including the nature of the causal variant(s) and the causal gene(s), as well as the directionality of effect. There are several tools that can be used for investigation of the functional genomics of these loci, and progress has been made on a limited number of novel CAD loci. New biology regarding atherosclerosis and CAD will be learned through the functional genomics of these loci, and the hope is that at least some of these new pathways relevant to CAD pathogenesis will yield new therapeutic targets for the prevention and treatment of CAD.
Collapse
Affiliation(s)
- Sylvia T Nurnberg
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Hanrui Zhang
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Nicholas J Hand
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Robert C Bauer
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Danish Saleheen
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Muredach P Reilly
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| | - Daniel J Rader
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| |
Collapse
|
66
|
Assimes TL, Lee IT, Juang JM, Guo X, Wang TD, Kim ET, Lee WJ, Absher D, Chiu YF, Hsu CC, Chuang LM, Quertermous T, Hsiung CA, Rotter JI, Sheu WHH, Chen YDI, Taylor KD. Genetics of Coronary Artery Disease in Taiwan: A Cardiometabochip Study by the Taichi Consortium. PLoS One 2016; 11:e0138014. [PMID: 26982883 PMCID: PMC4794124 DOI: 10.1371/journal.pone.0138014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/24/2015] [Indexed: 01/12/2023] Open
Abstract
By means of a combination of genome-wide and follow-up studies, recent large-scale association studies of populations of European descent have now identified over 46 loci associated with coronary artery disease (CAD). As part of the TAICHI Consortium, we have collected and genotyped 8556 subjects from Taiwan, comprising 5423 controls and 3133 cases with coronary artery disease, for 9087 CAD SNPs using the CardioMetaboChip. We applied penalized logistic regression to ascertain the top SNPs that contribute together to CAD susceptibility in Taiwan. We observed that the 9p21 locus contributes to CAD at the level of genome-wide significance (rs1537372, with the presence of C, the major allele, the effect estimate is -0.216, standard error 0.033, p value 5.8x10-10). In contrast to a previous report, we propose that the 9p21 locus is a single genetic contribution to CAD in Taiwan because: 1) the penalized logistic regression and the follow-up conditional analysis suggested that rs1537372 accounts for all of the CAD association in 9p21, and 2) the high linkage disequilibrium observed for all associated SNPs in 9p21. We also observed evidence for the following loci at a false discovery rate >5%: SH2B3, ADAMTS7, PHACTR1, GGCX, HTRA1, COL4A1, and LARP6-LRRC49. We also took advantage of the fact that penalized methods are an efficient approach to search for gene-by-gene interactions, and observed that two-way interactions between the PHACTR1 and ADAMTS7 loci and between the SH2B3 and COL4A1 loci contribute to CAD risk. Both the similarities and differences between the significance of these loci when compared with significance of loci in studies of populations of European descent underscore the fact that further genetic association of studies in additional populations will provide clues to identify the genetic architecture of CAD across all populations worldwide.
Collapse
Affiliation(s)
- Themistocles L. Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - I. -T. Lee
- Division of Endocrine and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jyh-Ming Juang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National University College of Medicine, Taipei, Taiwan
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, and Department of Pediatrics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tzung-Dau Wang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National University College of Medicine, Taipei, Taiwan
| | - Eric T. Kim
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, and Department of Pediatrics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - Yen-Feng Chiu
- Institute of Population Health Sciences, Division of Biostatistics and Bioinformatics, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Chih-Cheng Hsu
- Institute of Population Health Sciences, Division of Biostatistics and Bioinformatics, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Lee-Ming Chuang
- Division of Endocrine and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Thomas Quertermous
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Chao A. Hsiung
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, and Department of Pediatrics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Wayne H.-H. Sheu
- Division of Endocrine and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, and Department of Pediatrics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Kent D. Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, and Department of Pediatrics, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
67
|
Shared genetic aetiology of coronary artery disease and atherosclerotic stroke - 2015. Curr Atheroscler Rep 2015; 17:498. [PMID: 25690589 DOI: 10.1007/s11883-015-0498-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In the last years, genome-wide association studies have allowed to identify multiple genetic variants associated with atherosclerosis. In this review, we highlight the identification of genomic variants associated with coronary artery disease and myocardial infarction as well as large-vessel stroke. We will focus on genetic variants that displayed overlap for these atherosclerotic diseases. Current research is focusing on the identification of the functional mechanisms underlying these associations. As frequent variants are often only associated with small increases in risk, the search for the identification of rare variants with large increases in risk is ongoing. Whole-exome sequencing recently revealed rare variants dramatically increasing cardiovascular risk. Taken together, the developments of the past few years light the vision of improved prevention and therapy of coronary artery disease and stroke.
Collapse
|
68
|
Zhang P, Shen M, Fernandez-Patron C, Kassiri Z. ADAMs family and relatives in cardiovascular physiology and pathology. J Mol Cell Cardiol 2015; 93:186-99. [PMID: 26522853 DOI: 10.1016/j.yjmcc.2015.10.031] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/26/2015] [Accepted: 10/28/2015] [Indexed: 12/21/2022]
Abstract
A disintegrin and metalloproteinases (ADAMs) are a family of membrane-bound proteases. ADAM-TSs (ADAMs with thrombospondin domains) are a close relative of ADAMs that are present in soluble form in the extracellular space. Dysregulated production or function of these enzymes has been associated with pathologies such as cancer, asthma, Alzheimer's and cardiovascular diseases. ADAMs contribute to angiogenesis, hypertrophy and apoptosis in a stimulus- and cell type-dependent manner. Among the ADAMs identified so far (34 in mouse, 21 in human), ADAMs 8, 9, 10, 12, 17 and 19 have been shown to be involved in cardiovascular development or cardiomyopathies; and among the 19 ADAM-TSs, ADAM-TS1, 5, 7 and 9 are important in development of the cardiovascular system, while ADAM-TS13 can contribute to vascular disorders. Meanwhile, there remain a number of ADAMs and ADAM-TSs whose function in the cardiovascular system has not been yet explored. The current knowledge about the role of ADAMs and ADAM-TSs in the cardiovascular pathologies is still quite limited. The most detailed studies have been performed in other cell types (e.g. cancer cells) and organs (nervous system) which can provide valuable insight into the potential functions of ADAMs and ADAM-TSs, their mechanism of action and therapeutic potentials in cardiomyopathies. Here, we review what is currently known about the structure and function of ADAMs and ADAM-TSs, and their roles in development, physiology and pathology of the cardiovascular system.
Collapse
Affiliation(s)
- Pu Zhang
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Mengcheng Shen
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Carlos Fernandez-Patron
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
69
|
ADAMTS7 locus confers high cross-race risk for development of coronary atheromatous plaque. Mol Genet Genomics 2015; 291:121-8. [PMID: 26189211 DOI: 10.1007/s00438-015-1092-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 07/08/2015] [Indexed: 12/17/2022]
Abstract
Genome-wide association studies of coronary artery disease (CAD) have recently identified a new susceptibility locus, ADAMTS7, in subjects of European ancestry. However, the significance of this locus in Chinese populations has not been identified. Therefore, this study was designed to evaluate the effect of rs3825807, a non-synonymous variant in the prodomain of the ADAMTS7 protease, on CAD risk and atherosclerosis severity in a Chinese population. We performed genetic association analyses in two independent case-control cohorts, which included a total of 8154 participants. Additionally, the association between the ADAMTS7 rs3825807 genotype and the proportion of CAD patients with 3- and 1-vessel disease was tested. We found that ADAMTS7 rs3825807 was associated with susceptibility to CAD in a Chinese population [odds ratio (OR) = 1.15, 95 % confidence interval (CI) = 1.05-1.26, P = 0.002]. The association remained significant after adjusting for clinical covariates (adjusted OR = 1.12, 95 % CI = 1.02-1.24, P = 0.02). Among 3741 angiographically documented CAD patients, the rs3825807 risk allele showed a significant association with disease severity (P = 0.04, trend P = 0.02). Additionally, 3-vessel disease demonstrated a strong and direct association with ADAMTS7 rs3825807 gene dosage (P = 0.02). Overall, our findings indicate that the significant associations observed between this coding variant in ADAMTS7 and the risk of CAD development are cross-ethnic, and the gene dosage is consistent with the degree of coronary atheromatous burden.
Collapse
|
70
|
Kelwick R, Desanlis I, Wheeler GN, Edwards DR. The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin motifs) family. Genome Biol 2015; 16:113. [PMID: 26025392 PMCID: PMC4448532 DOI: 10.1186/s13059-015-0676-3] [Citation(s) in RCA: 424] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin motifs) enzymes are secreted, multi-domain matrix-associated zinc metalloendopeptidases that have diverse roles in tissue morphogenesis and patho-physiological remodeling, in inflammation and in vascular biology. The human family includes 19 members that can be sub-grouped on the basis of their known substrates, namely the aggrecanases or proteoglycanases (ADAMTS1, 4, 5, 8, 9, 15 and 20), the procollagen N-propeptidases (ADAMTS2, 3 and 14), the cartilage oligomeric matrix protein-cleaving enzymes (ADAMTS7 and 12), the von-Willebrand Factor proteinase (ADAMTS13) and a group of orphan enzymes (ADAMTS6, 10, 16, 17, 18 and 19). Control of the structure and function of the extracellular matrix (ECM) is a central theme of the biology of the ADAMTS, as exemplified by the actions of the procollagen-N-propeptidases in collagen fibril assembly and of the aggrecanases in the cleavage or modification of ECM proteoglycans. Defects in certain family members give rise to inherited genetic disorders, while the aberrant expression or function of others is associated with arthritis, cancer and cardiovascular disease. In particular, ADAMTS4 and 5 have emerged as therapeutic targets in arthritis. Multiple ADAMTSs from different sub-groupings exert either positive or negative effects on tumorigenesis and metastasis, with both metalloproteinase-dependent and -independent actions known to occur. The basic ADAMTS structure comprises a metalloproteinase catalytic domain and a carboxy-terminal ancillary domain, the latter determining substrate specificity and the localization of the protease and its interaction partners; ancillary domains probably also have independent biological functions. Focusing primarily on the aggrecanases and proteoglycanases, this review provides a perspective on the evolution of the ADAMTS family, their links with developmental and disease mechanisms, and key questions for the future.
Collapse
Affiliation(s)
- Richard Kelwick
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| | - Ines Desanlis
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| | - Grant N Wheeler
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| | - Dylan R Edwards
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
71
|
Zhang L, Yu F, Wang L, Zheng J, Du Y, Huang Y, Liu B, Wang X, Kong W. ADAMTS-7 promotes vascular smooth muscle cells proliferation in vitro and in vivo. SCIENCE CHINA-LIFE SCIENCES 2015; 58:674-81. [PMID: 25921940 DOI: 10.1007/s11427-015-4843-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/11/2015] [Indexed: 10/23/2022]
Abstract
Vascular smooth muscle cell (VSMC) proliferation and migration are pivotal for the pathogenesis of atherosclerosis and post-angioplasty restenosis. We have recently reported that a disintegrin and metalloproteinase with thrombospondin motifs-7 (ADAMTS-7), a novel metalloproteinase, contributes directly to neointima formation by mediating VSMC migration. However, whether ADAMTS-7 affects VSMC proliferation remains unclear. In this study, we found that luminal adenoviral delivery of ADAMTS-7 aggravated intimal hyperplasia 7 d after injury, paralleled by an increased percentage of PCNA-positive cells in both intima and media. In contrast, perivascular administration of ADAMTS-7 siRNA, but not scrambled siRNA to injured arteries attenuated intimal thickening at day 7, paralleled with reduced intimal VSMC replication, without alteration of VSMC proliferation in the media. In accordance, [(3)H]-thymidine incorporation assay in primary cultured rat VSMCs revealed an enhanced replication rate (by 61%) upon ADAMTS-7 overexpression and retarded proliferation (by 23%) upon ADAMTS-7 siRNA administration. Our data demonstrates that ADAMTS-7 promotes VSMC proliferation both in vitro and in vivo. ADAMTS-7 may therefore serve as a novel therapeutic target for atherosclerosis and post-angioplasty restenosis.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
ADAMTS-7 Inhibits Re-endothelialization of Injured Arteries and Promotes Vascular Remodeling Through Cleavage of Thrombospondin-1. Circulation 2015; 131:1191-201. [DOI: 10.1161/circulationaha.114.014072] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background—
ADAMTS-7, a member of the disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family, was recently identified to be significantly associated genomewide with coronary artery disease. However, the mechanisms that link ADAMTS-7 and coronary artery disease risk remain elusive. We have previously demonstrated that ADAMTS-7 promotes vascular smooth muscle cell migration and postinjury neointima formation via degradation of a matrix protein cartilage oligomeric matrix protein. Because delayed endothelium repair renders neointima and atherosclerosis plaque formation after vessel injury, we examined whether ADAMTS-7 also inhibits re-endothelialization.
Methods and Results—
Wire injury of the carotid artery and Evans blue staining were performed in
Adamts7
–/–
and wild-type mice. Adamts-7 deficiency greatly promoted re-endothelialization at 3, 5, and 7 days after injury. Consequently, Adamts-7 deficiency substantially ameliorated neointima formation in mice at days 14 and 28 after injury in comparison with the wild type. In vitro studies further indicated that ADAMTS-7 inhibited both endothelial cell proliferation and migration. Surprisingly, cartilage oligomeric matrix protein deficiency did not affect endothelial cell proliferation/migration and re-endothelialization in mice. In a further examination of other potential vascular substrates of ADAMTS-7, a label-free liquid chromatography-tandem mass spectrometry secretome analysis revealed thrombospondin-1 as a potential ADAMTS-7 target. The subsequent studies showed that ADAMTS-7 was directly associated with thrombospondin-1 by its C terminus and degraded thrombospondin-1 in vivo and in vitro. The inhibitory effect of ADAMTS-7 on postinjury endothelium recovery was circumvented in
Tsp1
–/–
mice.
Conclusions—
Our study revealed a novel mechanism by which ADAMTS-7 affects neointima formation. Thus, ADAMTS-7 is a promising treatment target for postinjury vascular intima hyperplasia.
Collapse
|
73
|
Dubail J, Apte SS. Insights on ADAMTS proteases and ADAMTS-like proteins from mammalian genetics. Matrix Biol 2015; 44-46:24-37. [PMID: 25770910 DOI: 10.1016/j.matbio.2015.03.001] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 01/05/2023]
Abstract
The mammalian ADAMTS superfamily comprises 19 secreted metalloproteinases and 7 ADAMTS-like proteins, each the product of a distinct gene. Thus far, all appear to be relevant to extracellular matrix function or to cell-matrix interactions. Most ADAMTS functions first emerged from analysis of spontaneous human and animal mutations and genetically engineered animals. The clinical manifestations of Mendelian disorders resulting from mutations in ADAMTS2, ADAMTS10, ADAMTS13, ADAMTS17, ADAMTSL2 and ADAMTSL4 identified essential roles for each gene, but also suggested potential cooperative functions of ADAMTS proteins. These observations were extended by analysis of spontaneous animal mutations, such as in bovine ADAMTS2, canine ADAMTS10, ADAMTS17 and ADAMTSL2 and mouse ADAMTS20. These human and animal disorders are recessive and their manifestations appear to result from a loss-of-function mechanism. Genome-wide analyses have determined an association of some ADAMTS loci such as ADAMTS9 and ADAMTS7, with specific traits and acquired disorders. Analysis of genetically engineered rodent mutations, now achieved for over half the superfamily, has provided novel biological insights and animal models for the respective human genetic disorders and suggested potential candidate genes for related human phenotypes. Engineered mouse mutants have been interbred to generate combinatorial mutants, uncovering cooperative functions of ADAMTS proteins in morphogenesis. Specific genetic models have provided crucial insights on mechanisms of osteoarthritis (OA), a common adult-onset degenerative condition. Engineered mutants will facilitate interpretation of exome variants identified in isolated birth defects and rare genetic conditions, as well as in genome-wide screens for trait and disease associations. Mammalian forward and reverse genetics, together with genome-wide analysis, together constitute a powerful force for revealing the functions of ADAMTS proteins in physiological pathways and health disorders. Their continuing use, together with genome-editing technology and the ability to generate stem cells from mutants, presents numerous opportunities for advancing basic knowledge, human disease pathways and therapy.
Collapse
Affiliation(s)
- Johanne Dubail
- Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Suneel S Apte
- Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
| |
Collapse
|
74
|
Zhang Y, Wei F, Liu CJ. Overexpression of ADAMTS-7 leads to accelerated initiation and progression of collagen-induced arthritis in mice. Mol Cell Biochem 2015; 404:171-9. [PMID: 25742929 DOI: 10.1007/s11010-015-2376-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/25/2015] [Indexed: 11/24/2022]
Abstract
The aim of the present study is to determine whether ADAMTS-7 contributes to the onset and severity of joint inflammation in the pathogenesis of inflammatory arthritis. ADAMTS-7 was found to be elevated in the course of collagen-induced arthritis (CIA). ADAMTS-7 transgenic (TG) mice were more susceptible to the induction of CIA. The onset of CIA was accelerated and the arthritic severity was increased in TG mice compared to wild-type mice. The overall incidence was also significantly higher in TG mice. In addition, arthritic TG mice displayed significantly higher clinical and histological arthritis scores. The COMP degradative fragments were significantly elevated in articular cartilage and sera in CIA models of TG mice. Furthermore, the production of tumor necrosis factor-alpha and interleukin-17 was also increased in serum and draining lymph nodes of arthritic TG mice. Therefore, these data provided the in vivo evidence, suggesting that ADAMTS-7 may play an important role in the pathogenesis of inflammatory arthritis, and that inhibition of ADAMTS-7 may be a potential target to ameliorate the severity of inflammatory arthritis.
Collapse
Affiliation(s)
- Yuying Zhang
- Department of Orthopaedic Surgery, New York University Medical Center, Rm 1608, HJD, 301 East 17th Street, New York, NY, 10003, USA
| | | | | |
Collapse
|
75
|
Affiliation(s)
- Alicia G Arroyo
- From Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Vicente Andrés
- From Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.
| |
Collapse
|
76
|
Bauer RC, Tohyama J, Cui J, Cheng L, Yang J, Zhang X, Ou K, Paschos GK, Zheng XL, Parmacek MS, Rader DJ, Reilly MP. Knockout of Adamts7, a novel coronary artery disease locus in humans, reduces atherosclerosis in mice. Circulation 2015; 131:1202-1213. [PMID: 25712206 DOI: 10.1161/circulationaha.114.012669] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Genome-wide association studies have established ADAMTS7 as a locus for coronary artery disease in humans. However, these studies fail to provide directionality for the association between ADAMTS7 and coronary artery disease. Previous reports have implicated ADAMTS7 in the regulation of vascular smooth muscle cell migration, but a role for and the direction of impact of this gene in atherogenesis have not been shown in relevant model systems. METHODS AND RESULTS We bred an Adamts7 whole-body knockout mouse onto both the Ldlr and Apoe knockout hyperlipidemic mouse models. Adamts7(-/-)/Ldlr(-/-) and Adamts7(-/-)/Apoe(-/-) mice displayed significant reductions in lesion formation in aortas and aortic roots compared with controls. Adamts7 knockout mice also showed reduced neointimal formation after femoral wire injury. Adamts7 expression was induced in response to injury and hyperlipidemia but was absent at later time points, and primary Adamts7 knockout vascular smooth muscle cells showed reduced migration in the setting of tumor necrosis factor-α stimulation. ADAMTS7 localized to cells positive for smooth muscle cell markers in human coronary artery disease lesions, and subcellular localization studies in cultured vascular smooth muscle cells placed ADAMTS7 at the cytoplasm and cell membrane, where it colocalized with markers of podosomes. CONCLUSIONS These data represent the first in vivo experimental validation of the association of Adamts7 with atherogenesis, likely through modulation of vascular cell migration and matrix in atherosclerotic lesions. These results demonstrate that Adamts7 is proatherogenic, lending directionality to the original genetic association and supporting the concept that pharmacological inhibition of ADAMTS7 should be atheroprotective in humans, making it an attractive target for novel therapeutic interventions.
Collapse
Affiliation(s)
- Robert C Bauer
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,Division of Translational Medicine and Human Genetics, Philadelphia, PA
| | - Junichiro Tohyama
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,Division of Translational Medicine and Human Genetics, Philadelphia, PA
| | - Jian Cui
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,The Cardiovascular Institute, Philadelphia, PA
| | - Lan Cheng
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,The Cardiovascular Institute, Philadelphia, PA
| | - Jifu Yang
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,The Cardiovascular Institute, Philadelphia, PA
| | - Xuan Zhang
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,The Cardiovascular Institute, Philadelphia, PA
| | - Kristy Ou
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,The Cardiovascular Institute, Philadelphia, PA
| | - Georgios K Paschos
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,The Institute of Translational Medicine and Therapeutics, Philadelphia, PA
| | - X Long Zheng
- The Children's Hospital of Philadelphia Department of Pathology and Laboratory Medicine, Philadelphia, PA
| | - Michael S Parmacek
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,The Cardiovascular Institute, Philadelphia, PA
| | - Daniel J Rader
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,Division of Translational Medicine and Human Genetics, Philadelphia, PA.,The Cardiovascular Institute, Philadelphia, PA.,The Institute of Translational Medicine and Therapeutics, Philadelphia, PA
| | - Muredach P Reilly
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,The Cardiovascular Institute, Philadelphia, PA
| |
Collapse
|
77
|
Kessler T, Kaess B, Bourier F, Erdmann J, Schunkert H. [Genetic analyses as basis for a personalized medicine in patients with coronary artery disease]. Herz 2014; 39:186-93. [PMID: 24464254 DOI: 10.1007/s00059-013-4048-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Knowledge about the etiology of coronary artery disease (CAD) entered new dimensions using genome-wide association studies. The current situation is that 46 chromosomal loci have been identified to be associated with CAD with genome-wide significance, i.e. p<5×10(-8), in Western Europeans. As the individual DNA sequence remains unchanged after fertilization, the risk variants cannot occur due to confounders, such as secondary disease processes. Thus, it can be proposed that these variants are directly affecting a primary and thereby causal pathophysiological process in CAD. Interestingly, only 20% of the effects mediated by the identified loci can be explained by the influence of traditional risk factors. This implies that yet unknown mechanisms and, as a consequence, new therapeutic targets play an important role in the pathophysiology of CAD. However, the high allele frequency of risk loci was also surprising. In the diploid chromosome set Western European individuals carry on average 30-50 risk variants at the 46 loci. Considering this, every individual in the population carries a larger or smaller genetic predisposition for CAD. On the other hand it is remarkable that many risk allele carriers seem to be able to compensate the genetic risk: even in old age not everyone suffers from CAD. This indicates yet unknown gene-gene and gene-environment interactions and limits the current possibilities in individual risk prediction.
Collapse
Affiliation(s)
- T Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Lazarettstr. 36, 80636, München, Deutschland
| | | | | | | | | |
Collapse
|
78
|
Abstract
PURPOSE OF REVIEW To review progress over the past 5 years in relating extracellular proteinases to plaque rupture, the cause of most myocardial infarctions, and consider the most promising prospects for developing related treatments. RECENT FINDINGS Cysteinyl cathepsins have been implicated in multiple macrophage functions that could promote plaque rupture. Cathepsin K is an attractive target because it is a collagenase and selective inhibitors are already being used in phase III clinical trials. Several serine proteinases clearly influence vascular remodelling and atherogenesis but important, unrelated actions limit their value as therapeutic targets. Among the metalloproteinases, new evidence supports roles for A Disintigrin and Metalloproteinases (ADAMs), including ADAM-10, ADAM-17 and ADAM-33, which suggest that selective inhibitors might be effective treatments. For ADAMs with ThromboSpondin domains (ADAMTSs), there are biological and genome-wide association data linking ADAMTS-7 to incidence of coronary heart disease but not increased risk of myocardial infarctions. In the case of matrix metalloproteinases (MMPs), selective inhibitors of MMP-12 and MMP-13 are available and may be appropriate for development as therapies. Novel targets, including MMP-8, MMP-10, MMP-14, MMP-19, MMP-25 and MMP-28, are also being considered. SUMMARY New opportunities exist to exploit proteinases as therapeutic targets in plaque rupture.
Collapse
Affiliation(s)
- Andrew C Newby
- University of Bristol and Bristol Heart Institute, Bristol, UK
| |
Collapse
|
79
|
Cartilage oligomeric matrix protein (COMP) in murine brachiocephalic and carotid atherosclerotic lesions. Atherosclerosis 2014; 236:366-72. [PMID: 25133350 PMCID: PMC4181795 DOI: 10.1016/j.atherosclerosis.2014.07.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 07/23/2014] [Accepted: 07/23/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To investigate the hypothesis that COMP can influence the morphology, stability and size of murine atherosclerotic lesions. METHODS ApoE- and ApoE/COMP-knockout mice were fed a high-fat diet to develop atherosclerotic plaques at lesion sites of three different types; inflammatory and fibrous plaques induced in the carotid artery by low or oscillatory shear stress, respectively, and spontaneously developing plaques in the brachiocephalic artery. The localization of COMP in the plaques and the effect of COMP deficiency on plaque development were evaluated. RESULTS COMP immunoreactivity was observed in about half of the investigated plaques from the ApoE null mice, mainly located along the intima-medial border. There were no significant differences in the size of inflammatory and fibrous carotid plaques between the genotypes. Plaques in the brachiocephalic artery from ApoE mice lacking COMP were increased in size with 54%. In these plaques the collagen content was also increased by 48%. There were no differences in relative collagen content in inflammatory and fibrous carotid plaques between genotypes. Polarized light microscopy showed that the increase in total collagen in brachiocephalic plaques was more than proportionally accounted for by an increase in thicker collagen fibrils. CONCLUSION We have shown that COMP deficiency has a significant impact on atherosclerotic plaque morphology and size. Our data also suggest that an altered collagen metabolism may be an important mechanism in this finding.
Collapse
|
80
|
Gao YX, Yu CA, Lu JH, Gao HM, Li G, Kong W, Zheng J. ADAMTS-7 expression increases in the early stage of angiotensin II-induced renal injury in elderly mice. Kidney Blood Press Res 2014; 38:121-31. [PMID: 24642842 DOI: 10.1159/000355758] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS We investigated the recently described family of proteinases, a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTs), and matrix metalloproteinases (MMPs) as inflammatory mediators in inflammatory kidney damage by studying ADAMTS-1, -4, and -7 and MMP-9 expression in elderly mouse kidneys after angiotensin II (Ang II) administration. METHODS Ang II (2.5 µg/kg/min) or norepinephrine (8.3 µg/kg/min) was subcutaneously infused in old mice. Renal injury was assessed by hematoxylin-eosin staining, 24-h albuminuria, and immunohistochemistry to evaluate inflammatory cell markers. The mRNA and protein expression of ADAMTS-1, -4, and -7 and MMP-9 were determined using real-time PCR, Western blot, and immunohistochemistry 3 days after Ang II or norepinephrine administration. RESULTS Elderly mice in the Ang II group developed hypertension and pathological kidney damage. The mRNA and protein levels of ADAMTS-7 in the Ang II group were 3.3 ± 1.1 (P = 0.019) and 1.6 ± 0.1 (P = 0.047) vs. 1.0 ± 0.1 and 1.0 ± 0.1 in the control group on day 3. In contrast, treatment with the hypertensive agent norepinephrine did not lead to obvious renal damage or an increase in renal ADAMTS-7 expression. CONCLUSIONS Renal ADAMTS-7 expression was induced by Ang II in elderly mice. The overexpression of ADATMTS-7 might contribute to early inflammatory kidney damage associated with aging.
Collapse
Affiliation(s)
- Yan-Xiang Gao
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, P. R. China
| | | | | | | | | | | | | |
Collapse
|
81
|
Wu C, Yan H, Sun J, Yang F, Song C, Jiang F, Li Y, Dong J, Zheng GY, Tian XL, Cao H. NEXN is a novel susceptibility gene for coronary artery disease in Han Chinese. PLoS One 2013; 8:e82135. [PMID: 24349201 PMCID: PMC3859596 DOI: 10.1371/journal.pone.0082135] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/21/2013] [Indexed: 01/14/2023] Open
Abstract
Coronary artery disease (CAD) is the leading cause of death and disability in the world. Genome-wide association studies have implicated the importance of the genetic contribution of vascular smooth muscle cells (VSMCs) function in CAD susceptibility. The aberrant phenotypic modulation of VSMC is responsible for the pathological vascular intima hyperplasia that is the hallmark for atherosclerotic morphology. NEXN is a muscle-specific F-actin binding protein and is regulated by inflammatory cytokines in VSMCs. Whether NEXN contributes to human vascular disorders is still unknown. In this study, we genotyped 5 SNPs, tagging all of the 17 common SNPs within 54 kilobases (kb) covering NEXN gene and its flanking region, in 1883 patients with CAD and 1973 healthy individuals from Han Chinese, and identified one SNP, rs1780050, which was strongly associated with CAD trait. The Bonferroni corrected P-value was 7.65×10(-5). The odds ratio (95% confidence interval) was 1.23 (1.12-1.36) with statistical power of 0.994. Functional analysis showed that NEXN promotes VSMC to a contractile phenotype in vitro and inhibits balloon-injury induced neointima formation in vivo. Further eQTL analysis demonstrated that the risk allele T of rs1780050 is associated with decreased expression of NEXN, thus contributing to a higher risk of CAD susceptibility in the population. This is, to our knowledge, the first study to identify NEXN as a novel CAD susceptibility gene with both genetic and functional evidence.
Collapse
Affiliation(s)
- Chong Wu
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Han Yan
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Jingzhi Sun
- Department of Cardiolody, Affiliated Hospital of Jining Medical University, Jining, China
| | - Fan Yang
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Chun Song
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Feng Jiang
- Department of Cardiology, Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yang Li
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Jie Dong
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Gu-Yan Zheng
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiao-Li Tian
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, China
- * E-mail: (XLT); (HC)
| | - Huiqing Cao
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, China
- * E-mail: (XLT); (HC)
| |
Collapse
|
82
|
Lai Y, Bai X, Zhao Y, Tian Q, Liu B, Lin EA, Chen Y, Lee B, Appleton CT, Beier F, Yu XP, Liu CJ. ADAMTS-7 forms a positive feedback loop with TNF-α in the pathogenesis of osteoarthritis. Ann Rheum Dis 2013; 73:1575-84. [PMID: 23928557 DOI: 10.1136/annrheumdis-2013-203561] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To examine the expression of ADAMTS-7 during the progression of osteoarthritis (OA), defining its role in the pathogenesis of OA, and elucidating the molecular events involved. METHODS ADAMTS-7 expression in cartilage of a rat OA model was assayed using immunohistochemistry. Cartilage-specific ADAMTS-7 transgenic mice and ADAMTS-7 small interfering (si)RNA knockdown mice were generated and used to analyse OA progression in both spontaneous and surgically induced OA models. Cartilage degradation and OA was evaluated using Safranin-O staining, immunohistochemistry, ELISA and western blotting. In addition, mRNA expression of tumour necrosis factor (TNF)-α and metalloproteinases known to be involved in cartilage degeneration in OA was analysed. Furthermore, the transactivation of ADAMTS-7 by TNF-α and its downstream NF-κB signalling was measured using reporter gene assay. RESULTS ADAMTS-7 expression was elevated during disease progression in the surgically induced rat OA model. Targeted overexpression of ADAMTS-7 in chondrocytes led to chondrodysplasia characterised by short-limbed dwarfism and a delay in endochondral ossification in 'young mice' and a spontaneous OA-like phenotype in 'aged' mice. In addition, overexpression of ADAMTS-7 led to exaggerated breakdown of cartilage and accelerated OA progression, while knockdown of ADAMTS-7 attenuated degradation of cartilage matrix and protected against OA development, in surgically induced OA models. ADAMTS-7 upregulated TNF-α and metalloproteinases associated with OA; in addition, TNF-α induced ADAMTS-7 through NF-κB signalling. CONCLUSIONS ADAMTS-7 and TNF-α form a positive feedback loop in the regulation of cartilage degradation and OA progression, making them potential molecular targets for prevention and treatment of joint degenerative diseases, including OA.
Collapse
Affiliation(s)
- Yongjie Lai
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York, USA Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xiaohui Bai
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York, USA
| | - Yunpeng Zhao
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York, USA
| | - Qingyun Tian
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York, USA
| | - Ben Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York, USA
| | - Edward A Lin
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York, USA
| | - Yuqing Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Howard Hughes Medical Institute, Houston, Texas, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Howard Hughes Medical Institute, Houston, Texas, USA
| | - C Thomas Appleton
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Alberta, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Alberta, Canada
| | - Xiu-Ping Yu
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Chuan-Ju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York, USA Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
83
|
Patel RS, Ye S. ADAMTS7: a promising new therapeutic target in coronary heart disease. Expert Opin Ther Targets 2013; 17:863-7. [PMID: 23829786 DOI: 10.1517/14728222.2013.816287] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Studies in animal models have demonstrated that the protease ADAMTS7 plays a role in neointima formation after arterial mechanical injury, by facilitating vascular smooth muscle cell (VSMC) migration. Furthermore, recent human genetic studies have revealed an association between DNA polymorphisms at the ADAMTS7 gene locus and risk of coronary artery disease (CAD). Functional studies have shown that a CAD-associated polymorphism in the coding region of the ADAMTS7 gene affects ADAMTS7 maturation and VSMC migration. This editorial highlights these findings and discusses targeted ADAMTS7 inhibition as a possible novel approach to treat CAD.
Collapse
|
84
|
Abstract
Here, we provide a comprehensive review of current findings concerning the biochemistry and physiological functions of ADAMTS7, a metalloprotease that is known to interact with cartilage oligomeric matrix protein, progranulin, and alpha2-macroglobulin. Such broad substrate specificity and potentially diverse physiological functions make ADAMTS7 an interesting enzyme to study. ADAMTS7 has been shown to play a role in the pathogenesis of arthritis and disc disorders. More recently, the ADAMTS7 locus is identified to have a strong association with coronary atherosclerotic disease. However, the role of ADAMTS7 in the development of atherosclerosis is yet to be determined. The development of an easy and high throughput assay for ADAMTS7 activity and appropriate animal models will allow us to uncover the novel mechanisms of coronary arterial disease.
Collapse
Affiliation(s)
- Hayley A Hanby
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | | |
Collapse
|