301
|
Yang Z, Zhang Q, Yu H, Du H, Li L, He Y, Zhu S, Li C, Zhang S, Luo B, Gao Y. Genetic association study of a novel indel polymorphism in HSPA1B with the risk of sudden cardiac death in the Chinese populations. Forensic Sci Int 2020; 318:110637. [PMID: 33309992 DOI: 10.1016/j.forsciint.2020.110637] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/16/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022]
Abstract
Sudden cardiac death (SCD) has become a global problem due to its high mortality in the general population. Identification of genetic factors predisposed to SCD is significant since it enables genetic testing that would contribute to molecular diagnosis and risk stratification of SCD. It has been reported that HSPA1B gene mutations might be related with SCD. In this study, based on candidate-gene-based approach and systematic screening strategy, a 5-base pair insertion/deletion (Indel) polymorphism (rs3036297) in the 3'UTR of HSPA1B gene was selected to perform a case-control study aiming to investigate its association with SCD susceptibility in Chinese populations. Logistic regression analysis showed that the insertion allele of rs3036297 was correlated with a comparatively lower risk for SCD [OR=0.58, 95%CI=0.43-0.77, P=1.28×10-4] compared with the deletion allele. Luciferase activity assay indicated that HSPA1B expression could be regulated by rs3036297 through interfering binding with miR-134-5p. Furthermore, analysis of database from Haploreg and GTEx revealed that the rs3036297 variant was involved in potential cis-regulatory element with the promoter of HLA-DRB5 through a long-range interaction and the deletion allele of rs3036297 increased HLA-DRB5 expression. In conclusion, the rs3036297 variant may regulate HSPA1B expression via a mechanism of miRNA binding and HLA-DRB5 expression via a long-range promoter interaction through which contributed to SCD susceptibility. Therefore, rs3036297 would be a potential marker for molecular diagnosis and genetic counseling of SCD.
Collapse
Affiliation(s)
- Zhenzhen Yang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China; Institute of Forensic Sciences, Henan University of Economics and Law, Zhengzhou, China
| | - Qing Zhang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China
| | - Huan Yu
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China
| | - Hailin Du
- Nanjing Red Cross Blood Center, Nanjing, China
| | - Lijuan Li
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China
| | - Yan He
- Department of Epidemiology, Medical College of Soochow University, Suzhou, China
| | - Shaohua Zhu
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China
| | - Chengtao Li
- Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Sciences, Ministry of Justice, Shanghai, China
| | - Suhua Zhang
- Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Sciences, Ministry of Justice, Shanghai, China
| | - Bin Luo
- Department of Forensic Pathology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Yuzhen Gao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
302
|
Surendran P, Feofanova EV, Lahrouchi N, Ntalla I, Karthikeyan S, Cook J, Chen L, Mifsud B, Yao C, Kraja AT, Cartwright JH, Hellwege JN, Giri A, Tragante V, Thorleifsson G, Liu DJ, Prins BP, Stewart ID, Cabrera CP, Eales JM, Akbarov A, Auer PL, Bielak LF, Bis JC, Braithwaite VS, Brody JA, Daw EW, Warren HR, Drenos F, Nielsen SF, Faul JD, Fauman EB, Fava C, Ferreira T, Foley CN, Franceschini N, Gao H, Giannakopoulou O, Giulianini F, Gudbjartsson DF, Guo X, Harris SE, Havulinna AS, Helgadottir A, Huffman JE, Hwang SJ, Kanoni S, Kontto J, Larson MG, Li-Gao R, Lindström J, Lotta LA, Lu Y, Luan J, Mahajan A, Malerba G, Masca NGD, Mei H, Menni C, Mook-Kanamori DO, Mosen-Ansorena D, Müller-Nurasyid M, Paré G, Paul DS, Perola M, Poveda A, Rauramaa R, Richard M, Richardson TG, Sepúlveda N, Sim X, Smith AV, Smith JA, Staley JR, Stanáková A, Sulem P, Thériault S, Thorsteinsdottir U, Trompet S, Varga TV, Velez Edwards DR, Veronesi G, Weiss S, Willems SM, Yao J, Young R, Yu B, Zhang W, Zhao JH, Zhao W, Zhao W, Evangelou E, Aeschbacher S, Asllanaj E, Blankenberg S, Bonnycastle LL, Bork-Jensen J, Brandslund I, Braund PS, Burgess S, et alSurendran P, Feofanova EV, Lahrouchi N, Ntalla I, Karthikeyan S, Cook J, Chen L, Mifsud B, Yao C, Kraja AT, Cartwright JH, Hellwege JN, Giri A, Tragante V, Thorleifsson G, Liu DJ, Prins BP, Stewart ID, Cabrera CP, Eales JM, Akbarov A, Auer PL, Bielak LF, Bis JC, Braithwaite VS, Brody JA, Daw EW, Warren HR, Drenos F, Nielsen SF, Faul JD, Fauman EB, Fava C, Ferreira T, Foley CN, Franceschini N, Gao H, Giannakopoulou O, Giulianini F, Gudbjartsson DF, Guo X, Harris SE, Havulinna AS, Helgadottir A, Huffman JE, Hwang SJ, Kanoni S, Kontto J, Larson MG, Li-Gao R, Lindström J, Lotta LA, Lu Y, Luan J, Mahajan A, Malerba G, Masca NGD, Mei H, Menni C, Mook-Kanamori DO, Mosen-Ansorena D, Müller-Nurasyid M, Paré G, Paul DS, Perola M, Poveda A, Rauramaa R, Richard M, Richardson TG, Sepúlveda N, Sim X, Smith AV, Smith JA, Staley JR, Stanáková A, Sulem P, Thériault S, Thorsteinsdottir U, Trompet S, Varga TV, Velez Edwards DR, Veronesi G, Weiss S, Willems SM, Yao J, Young R, Yu B, Zhang W, Zhao JH, Zhao W, Zhao W, Evangelou E, Aeschbacher S, Asllanaj E, Blankenberg S, Bonnycastle LL, Bork-Jensen J, Brandslund I, Braund PS, Burgess S, Cho K, Christensen C, Connell J, Mutsert RD, Dominiczak AF, Dörr M, Eiriksdottir G, Farmaki AE, Gaziano JM, Grarup N, Grove ML, Hallmans G, Hansen T, Have CT, Heiss G, Jørgensen ME, Jousilahti P, Kajantie E, Kamat M, Käräjämäki A, Karpe F, Koistinen HA, Kovesdy CP, Kuulasmaa K, Laatikainen T, Lannfelt L, Lee IT, Lee WJ, Linneberg A, Martin LW, Moitry M, Nadkarni G, Neville MJ, Palmer CNA, Papanicolaou GJ, Pedersen O, Peters J, Poulter N, Rasheed A, Rasmussen KL, Rayner NW, Mägi R, Renström F, Rettig R, Rossouw J, Schreiner PJ, Sever PS, Sigurdsson EL, Skaaby T, Sun YV, Sundstrom J, Thorgeirsson G, Esko T, Trabetti E, Tsao PS, Tuomi T, Turner ST, Tzoulaki I, Vaartjes I, Vergnaud AC, Willer CJ, Wilson PWF, Witte DR, Yonova-Doing E, Zhang H, Aliya N, Almgren P, Amouyel P, Asselbergs FW, Barnes MR, Blakemore AI, Boehnke M, Bots ML, Bottinger EP, Buring JE, Chambers JC, Chen YDI, Chowdhury R, Conen D, Correa A, Davey Smith G, Boer RAD, Deary IJ, Dedoussis G, Deloukas P, Di Angelantonio E, Elliott P, Felix SB, Ferrières J, Ford I, Fornage M, Franks PW, Franks S, Frossard P, Gambaro G, Gaunt TR, Groop L, Gudnason V, Harris TB, Hayward C, Hennig BJ, Herzig KH, Ingelsson E, Tuomilehto J, Järvelin MR, Jukema JW, Kardia SLR, Kee F, Kooner JS, Kooperberg C, Launer LJ, Lind L, Loos RJF, Majumder AAS, Laakso M, McCarthy MI, Melander O, Mohlke KL, Murray AD, Nordestgaard BG, Orho-Melander M, Packard CJ, Padmanabhan S, Palmas W, Polasek O, Porteous DJ, Prentice AM, Province MA, Relton CL, Rice K, Ridker PM, Rolandsson O, Rosendaal FR, Rotter JI, Rudan I, Salomaa V, Samani NJ, Sattar N, Sheu WHH, Smith BH, Soranzo N, Spector TD, Starr JM, Sebert S, Taylor KD, Lakka TA, Timpson NJ, Tobin MD, van der Harst P, van der Meer P, Ramachandran VS, Verweij N, Virtamo J, Völker U, Weir DR, Zeggini E, Charchar FJ, Wareham NJ, Langenberg C, Tomaszewski M, Butterworth AS, Caulfield MJ, Danesh J, Edwards TL, Holm H, Hung AM, Lindgren CM, Liu C, Manning AK, Morris AP, Morrison AC, O'Donnell CJ, Psaty BM, Saleheen D, Stefansson K, Boerwinkle E, Chasman DI, Levy D, Newton-Cheh C, Munroe PB, Howson JMM. Discovery of rare variants associated with blood pressure regulation through meta-analysis of 1.3 million individuals. Nat Genet 2020; 52:1314-1332. [PMID: 33230300 PMCID: PMC7610439 DOI: 10.1038/s41588-020-00713-x] [Show More Authors] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/08/2020] [Indexed: 01/14/2023]
Abstract
Genetic studies of blood pressure (BP) to date have mainly analyzed common variants (minor allele frequency > 0.05). In a meta-analysis of up to ~1.3 million participants, we discovered 106 new BP-associated genomic regions and 87 rare (minor allele frequency ≤ 0.01) variant BP associations (P < 5 × 10-8), of which 32 were in new BP-associated loci and 55 were independent BP-associated single-nucleotide variants within known BP-associated regions. Average effects of rare variants (44% coding) were ~8 times larger than common variant effects and indicate potential candidate causal genes at new and known loci (for example, GATA5 and PLCB3). BP-associated variants (including rare and common) were enriched in regions of active chromatin in fetal tissues, potentially linking fetal development with BP regulation in later life. Multivariable Mendelian randomization suggested possible inverse effects of elevated systolic and diastolic BP on large artery stroke. Our study demonstrates the utility of rare-variant analyses for identifying candidate genes and the results highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Elena V Feofanova
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Najim Lahrouchi
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences Amsterdam, Amsterdam, the Netherlands
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Savita Karthikeyan
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - James Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Lingyan Chen
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Borbala Mifsud
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Chen Yao
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - 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
| | - James H Cartwright
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jacklyn N Hellwege
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Ayush Giri
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
- Division of Quantitative Sciences, Department of Obstetrics & Gynecology, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Vinicius Tragante
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
| | | | - Dajiang J Liu
- Institute of Personalized Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Bram P Prins
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Isobel D Stewart
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - 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 Centre, Queen Mary University of London, London, UK
| | - James M Eales
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Artur Akbarov
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Paul L Auer
- Joseph J Zilber School of Public Health, University of Wisconsin, Milwaukee, WI, USA
| | - Lawrence F Bielak
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Vickie S Braithwaite
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
- MRC Nutrition and Bone Health Group, University of Cambridge, Cambridge, UK
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - E Warwick Daw
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - 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 Centre, Queen Mary University of London, London, UK
| | - Fotios Drenos
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
| | - Sune Fallgaard Nielsen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Eric B Fauman
- Internal Medicine Research Unit, Pfizer, Cambridge, MA, USA
| | - Cristiano Fava
- Department of Medicine, University of Verona, Verona, Italy
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Teresa Ferreira
- The Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Christopher N Foley
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - He Gao
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
| | - Olga Giannakopoulou
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Genomic Health, Queen Mary University of London, London, UK
- Division of Psychiatry, University College of London, London, UK
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Aki S Havulinna
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | | | - Jennifer E Huffman
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, UK
| | - Shih-Jen Hwang
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Boston University and National Heart, Lung and Blood Institute Framingham Heart Study, Framingham, MA, USA
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Genomic Health, Life Sciences, Queen Mary University of London, London, UK
| | - Jukka Kontto
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Martin G Larson
- Boston University and National Heart, Lung and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Biostatistics Department, Boston University School of Public Health, Boston, MA, USA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jaana Lindström
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Luca A Lotta
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Giovanni Malerba
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Nicholas G D Masca
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Hao Mei
- Department of Data Science, School of Population Health, University of Mississippi Medical Center, Jackson, MS, USA
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - David Mosen-Ansorena
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, Ludwig-Maximilians-University Munich, Munich, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU, Munich, Germany
| | - Guillaume Paré
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dirk S Paul
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Markus Perola
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- Clinical and Molecular Metabolism Research Program (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Alaitz Poveda
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Melissa Richard
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nuno Sepúlveda
- Department of Infection Biology, Faculty of Tropical and Infectious Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Centre of Statistics and Applications of University of Lisbon, Lisbon, Portugal
| | - Xueling Sim
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Saw Swee Hock School of Public Health, National University of, Singapore, Singapore
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - James R Staley
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Alena Stanáková
- University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | | | - Sébastien Thériault
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, Quebec, Canada
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Stella Trompet
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tibor V Varga
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Digna R Velez Edwards
- Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Tennessee Valley Health Systems VA, Nashville, TN, USA
| | - Giovanni Veronesi
- Research Center in Epidemiology and Preventive Medicine, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and University of Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Sara M Willems
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Robin Young
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Bing Yu
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
| | - Jing-Hua Zhao
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Wei Zhao
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | | | - Eralda Asllanaj
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Stefan Blankenberg
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
- University Medical Center Hamburg Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lori L Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ivan Brandslund
- Department of Clinical Biochemistry, Lillebaelt Hospital, Vejle, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Stephen Burgess
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - John Connell
- University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anna F Dominiczak
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | | | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
- Department of Population Science and Experimental Medicine, Institute of Cardiovascular Science, University College London, London, UK
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian T Have
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gerardo Heiss
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | | | - Pekka Jousilahti
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Eero Kajantie
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Hospital for Children and Adolescents, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Mihir Kamat
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - AnneMari Käräjämäki
- Department of Primary Health Care, Vaasa Central Hospital, Vaasa, Finland
- Diabetes Center, Vaasa Health Care Center, Vaasa, Finland
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
| | - Heikki A Koistinen
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Csaba P Kovesdy
- Nephrology Section, Memphis VA Medical Center, Memphis, TN, USA
| | - Kari Kuulasmaa
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Tiina Laatikainen
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Lars Lannfelt
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- School of Medicine, , Chung Shan Medical University, Taichung, Taiwan
- College of Science, Tunghai University, Taichung, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Social Work, Tunghai University, Taichung, Taiwan
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisa W Martin
- George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Marie Moitry
- Department of Public health, Strasbourg University Hospital, University of Strasbourg, Strasbourg, France
| | - Girish Nadkarni
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matt J Neville
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
| | - Colin N A Palmer
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, UK
| | | | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - James Peters
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Neil Poulter
- International Centre for Circulatory Health, Imperial College London, London, UK
| | - Asif Rasheed
- Centre for Non-Communicable Diseases, Karachi, Pakistan
| | - Katrine L Rasmussen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - N William Rayner
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Reedik Mägi
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Frida Renström
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Rainer Rettig
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Jacques Rossouw
- Division of Cardiovascular Sciences, NHLBI, Bethesda, MD, USA
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Peter S Sever
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Emil L Sigurdsson
- Department of Family Medicine, University of Iceland, Reykjavik, Iceland
- Development Centre for Primary Health Care in Iceland, Reykjavik, Iceland
| | - Tea Skaaby
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Johan Sundstrom
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Internal Medicine, Division of Cardiology, Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - Tõnu Esko
- Institute of Genomics, University of Tartu, Tartu, Estonia
- Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Elisabetta Trabetti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Philip S Tsao
- VA Palo Alto Health Care System, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tiinamaija Tuomi
- Folkhälsan Research Centre, Helsinki, Finland
- Department of Endocrinology, Helsinki University Central Hospital, Helsinki, Finland
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden Institute for Molecular Medicine Helsinki (FIMM), Helsinki University, Helsinki, Finland
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Ilonca Vaartjes
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University of Utrecht, University of Utrecht, Utrecht, the Netherlands
- Center for Circulatory Health, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Anne-Claire Vergnaud
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Peter W F Wilson
- Atlanta VAMC and Emory Clinical Cardiovascular Research Institute, Atlanta, GA, USA
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark
- Danish Diabetes Academy, Odense, Denmark
- Steno Diabetes Center Aarhus, Aarhus, Denmark
| | - Ekaterina Yonova-Doing
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - He Zhang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Naheed Aliya
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Peter Almgren
- Department of Medicine, Lund University, Malmö, Sweden
| | - Philippe Amouyel
- Univ Lille, U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, France
- INSERM, U1167, Lille, France
- CHU Lille, U1167, Lille, France
- Institut Pasteur de Lille, U1167, Lille, France
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
- Health Data Research UK, Institute of Health Informatics, University College London, London, 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 Centre, Queen Mary University of London, London, UK
| | - Alexandra I Blakemore
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
- Section of Investigative Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Michiel L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University of Utrecht, University of Utrecht, Utrecht, the Netherlands
- Center for Circulatory Health, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie E Buring
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - John C Chambers
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Imperial College Healthcare NHS Trust, London, UK
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Rajiv Chowdhury
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Non-communicable Disease Research (CNCR), Dhaka, Bangladesh
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Cardiovascular Research Institute Basel, Basel, Switzerland
| | - Adolfo Correa
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Rudolf A de Boer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Panos Deloukas
- 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 Centre, Queen Mary University of London, London, UK
- Centre for Genomic Health, Life Sciences, Queen Mary University of London, London, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit (BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Health Data Research UK-London at Imperial College London, London, UK
- UKDRI, Dementia Research Institute at Imperial College London, London, UK
- British Heart Foundation (BHF) Centre of Research Excellence, Imperial College London, London, UK
| | - Stephan B Felix
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Jean Ferrières
- Department of Cardiology and Department of Epidemiology, INSERM UMR 1027, Toulouse University Hospital, Toulouse, France
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Myriam Fornage
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Sweden
- Oxford Center for Diabetes, Endocrinology & Metabolism, Radcliff Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen Franks
- Institute of Reproductive & Developmental Biology, Imperial College London, London, UK
| | | | - Giovanni Gambaro
- Division of Nephrology, Department of Medicine, University of Verona, Verona, Italy
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Medicine Helsinki (FIMM), Helsinki University, Helsinki, Finland
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute of Aging, Bethesda, MD, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, UK
| | - Branwen J Hennig
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, Gambia
- Wellcome Trust, London, UK
| | - Karl-Heinz Herzig
- Institute of Biomedicine, Medical Research Center (MRC), University of Oulu, and University Hospital Oulu, Oulu, Finland
- Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Jaakko Tuomilehto
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Saudi Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
- National Institute of Public Health, Madrid, Spain
| | - Marjo-Riitta Järvelin
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Unit of Primary Care, Oulu University Hospital, Kajaanintie, Oulu, Finland
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Frank Kee
- Centre for Public Health, Queens University Belfast, Belfast, UK
| | - Jaspal S Kooner
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Charles Kooperberg
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, WA, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute of Aging, Bethesda, MD, USA
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- Genentech, South San Francisco, San Francisco, CA, USA
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Alison D Murray
- The Institute of Medical Sciences, Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Børge Grønne Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | | | | | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Walter Palmas
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Ozren Polasek
- Department of Public Health, University of Split School of Medicine, Split, Croatia
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Andrew M Prentice
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, Gambia
- MRC International Nutrition Group at London School of Hygiene and Tropical Medicine, Keppel St, London, UK
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Olov Rolandsson
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Sweden
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Scotland, UK
| | - Veikko Salomaa
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Wayne H-H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
- Institute of Medical Technology, National Chung-Hsing University, Taichung, Taiwan
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Nicole Soranzo
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit (BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Research Centre, University of Edinburgh, Edinburgh, UK
| | - Sylvain Sebert
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Timo A Lakka
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Institute of Biomedicine/Physiology, University of Eastern Finland, Kuopio, Finland
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Martin D Tobin
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands
| | - Peter van der Meer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Vasan S Ramachandran
- Boston University and National Heart, Lung and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - Niek Verweij
- University Medical Center Groningen, Groningen, the Netherlands
| | - Jarmo Virtamo
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and University of Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Eleftheria Zeggini
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Institute of Translational Genomics, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- TUM School of Medicine, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| | - Fadi J Charchar
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Health Innovation and Transformation Center, Federation University Australia, Ballarat, Victoria, Australia
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Division of Medicine, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit (BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge, 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 Centre, Queen Mary University of London, London, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit (BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Hilma Holm
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
| | - Adriana M Hung
- VA Tennessee Valley Healthcare System, Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cecilia M Lindgren
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- The Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Chunyu Liu
- Boston University School of Public Health, Boston, MA, USA
| | - Alisa K Manning
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester, UK
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christopher J O'Donnell
- VA Boston Healthcare, Section of Cardiology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Danish Saleheen
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Daniel Levy
- Boston University and National Heart, Lung and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Population Sciences, Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MD, USA
| | - Christopher Newton-Cheh
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - 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 Centre, Queen Mary University of London, London, UK.
| | - Joanna M M Howson
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK.
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK.
- Novo Nordisk Research Centre Oxford, Novo Nordisk Ltd, Oxford, UK.
| |
Collapse
|
303
|
Chen R, Morris BJ, Donlon TA, Masaki KH, Willcox DC, Davy PM, Allsopp RC, Willcox BJ. FOXO3 longevity genotype mitigates the increased mortality risk in men with a cardiometabolic disease. Aging (Albany NY) 2020; 12:23509-23524. [PMID: 33260156 PMCID: PMC7762472 DOI: 10.18632/aging.202175] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/22/2020] [Indexed: 12/31/2022]
Abstract
FOXO3 is a prominent longevity gene. To date, no-one has examined whether longevity-associated FOXO3 genetic variants protect against mortality in all individuals, or only in those with aging-related diseases. We therefore tested longevity-associated FOXO3 single nucleotide polymorphisms in a haplotype block for association with mortality in 3,584 elderly American men of Japanese ancestry, 2,512 with and 1,072 without a cardiometabolic disease (CMD). At baseline (1991-1993), 1,010 CMD subjects had diabetes, 1,919 had hypertension, and 738 had coronary heart disease (CHD). Follow-up until Dec 31, 2019 found that in CMD-affected individuals, longevity-associated alleles of FOXO3 were associated with significantly longer lifespan: haplotype hazard ratio 0.81 (95% CI 0.72-0.91; diabetes 0.77, hypertension 0.82, CHD 0.83). Overall, men with a CMD had higher mortality than men without a CMD (P=6x10-7). However, those men with a CMD who had the FOXO3 longevity genotype had similar survival as men without a CMD. In men without a CMD there was no association of longevity-associated alleles of FOXO3 with lifespan. Our study provides novel insights into the basis for the long-established role of FOXO3 as a longevity gene. We suggest that the FOXO3 longevity genotype increases lifespan only in at-risk individuals by protection against cardiometabolic stress.
Collapse
Affiliation(s)
- Randi Chen
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA
| | - Brian J. Morris
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Timothy A. Donlon
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
- Institute for Biogenesis Research, University of Hawaii, Honolulu, HI 96822, USA
| | - Kamal H. Masaki
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - D. Craig Willcox
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA
- Department of Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Philip M.C. Davy
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA
- Department of Human Welfare, Okinawa International University, Ginowan, Okinawa, Japan
| | - Richard C. Allsopp
- Department of Human Welfare, Okinawa International University, Ginowan, Okinawa, Japan
| | - Bradley J. Willcox
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| |
Collapse
|
304
|
van Zuydam NR, Ladenvall C, Voight BF, Strawbridge RJ, Fernandez-Tajes J, Rayner NW, Robertson NR, Mahajan A, Vlachopoulou E, Goel A, Kleber ME, Nelson CP, Kwee LC, Esko T, Mihailov E, Mägi R, Milani L, Fischer K, Kanoni S, Kumar J, Song C, Hartiala JA, Pedersen NL, Perola M, Gieger C, Peters A, Qu L, Willems SM, Doney AS, Morris AD, Zheng Y, Sesti G, Hu FB, Qi L, Laakso M, Thorsteinsdottir U, Grallert H, van Duijn C, Reilly MP, Ingelsson E, Deloukas P, Kathiresan S, Metspalu A, Shah SH, Sinisalo J, Salomaa V, Hamsten A, Samani NJ, März W, Hazen SL, Watkins H, Saleheen D, Morris AP, Colhoun HM, Groop L, McCarthy MI, Palmer CN. Genetic Predisposition to Coronary Artery Disease in Type 2 Diabetes Mellitus. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2020; 13:e002769. [PMID: 33321069 PMCID: PMC7748049 DOI: 10.1161/circgen.119.002769] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 07/01/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Coronary artery disease (CAD) is accelerated in subjects with type 2 diabetes mellitus (T2D). METHODS To test whether this reflects differential genetic influences on CAD risk in subjects with T2D, we performed a systematic assessment of genetic overlap between CAD and T2D in 66 643 subjects (27 708 with CAD and 24 259 with T2D). Variants showing apparent association with CAD in stratified analyses or evidence of interaction were evaluated in a further 117 787 subjects (16 694 with CAD and 11 537 with T2D). RESULTS None of the previously characterized CAD loci was found to have specific effects on CAD in T2D individuals, and a genome-wide interaction analysis found no new variants for CAD that could be considered T2D specific. When we considered the overall genetic correlations between CAD and its risk factors, we found no substantial differences in these relationships by T2D background. CONCLUSIONS This study found no evidence that the genetic architecture of CAD differs in those with T2D compared with those without T2D.
Collapse
Affiliation(s)
- Natalie R. van Zuydam
- Pat Macpherson Center for Pharmacogenetics & Pharmacogenomics, Cardiovascular & Diabetes Medicine (N.R.v.Z., C.N.A.P.), School of Medicine, University of Dundee
- Oxford Center for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine (N.R.v.Z., N.W.R., N.R.R., A. Mahajan, M.I.Mc), University of Oxford, United Kingdom
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
| | - Claes Ladenvall
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University Diabetes Center, Malmö, Sweden (C.L., L.G.)
| | - Benjamin F. Voight
- Department of Systems Pharmacology & Translational Therapeutics (B.F.V.)
- Department of Genetics (B.F.V.)
- Institute for Translational Medicine & Therapeutics (B.F.V.)
| | - Rona J. Strawbridge
- Cardiovascular Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden (R.J.S., A.H.)
| | - Juan Fernandez-Tajes
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
| | - N. William Rayner
- Oxford Center for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine (N.R.v.Z., N.W.R., N.R.R., A. Mahajan, M.I.Mc), University of Oxford, United Kingdom
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom (N.W.R.)
| | - Neil R. Robertson
- Oxford Center for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine (N.R.v.Z., N.W.R., N.R.R., A. Mahajan, M.I.Mc), University of Oxford, United Kingdom
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
| | - Anubha Mahajan
- Oxford Center for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine (N.R.v.Z., N.W.R., N.R.R., A. Mahajan, M.I.Mc), University of Oxford, United Kingdom
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
| | - Efthymia Vlachopoulou
- Transplantation Laboratory, Haartman Institute (E.V.), University of Helsinki, Helsinki, Finland
| | - Anuj Goel
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
- Division of Cardiovascular Medicine (A.G., H.W.), University of Oxford, United Kingdom
| | - Marcus E. Kleber
- Pat Macpherson Center for Pharmacogenetics & Pharmacogenomics, Cardiovascular & Diabetes Medicine (N.R.v.Z., C.N.A.P.), School of Medicine, University of Dundee
- Division of Molecular & Clinical Medicine (A.S.F.D.), School of Medicine, University of Dundee
- Oxford Center for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine (N.R.v.Z., N.W.R., N.R.R., A. Mahajan, M.I.Mc), University of Oxford, United Kingdom
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
- Division of Cardiovascular Medicine (A.G., H.W.), University of Oxford, United Kingdom
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University Diabetes Center, Malmö, Sweden (C.L., L.G.)
- Department of Systems Pharmacology & Translational Therapeutics (B.F.V.)
- Department of Genetics (B.F.V.)
- Institute for Translational Medicine & Therapeutics (B.F.V.)
- Cardiovascular Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA (L.Q., M.P.R.)
- Cardiovascular Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden (R.J.S., A.H.)
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom (N.W.R.)
- Transplantation Laboratory, Haartman Institute (E.V.), University of Helsinki, Helsinki, Finland
- Research Program for Clinical & Molecular Metabolism, Faculty of Medicine (M.P.), University of Helsinki, Helsinki, Finland. Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Cardiovascular Sciences, University of Leicester (C.P.N., N.J.S.)
- NIHR Leicester Biomedical Research Center, Glenfield Hospital, Leicester, United Kingdom (C.P.N., N.J.S.)
- Division of Cardiology, Department of Medicine, Duke University Medical Center (S.H.S.)
- Duke Molecular Physiology Institute, Duke University, Durham, NC (L.C.K., S.H.S.)
- Estonian Genome Center (T.E., E.M., R.M., L.M., K.F., M.P., A. Metspalu), University of Tartu, Tartu, Estonia
- Institute of Cell & Molecular Biology (A. Metspalu), University of Tartu, Tartu, Estonia
- Center for Genomic Health (S.K.), Queen Mary University of London, London, United Kingdom
- William Harvey Research Institute, Barts & the London Medical School (S.K., P.D.), Queen Mary University of London, London, United Kingdom
- Department of Medical Sciences, Molecular Epidemiology & Science for Life Laboratory (J.K., C.S., E.I.)
- Department of Immunology, Genetics and Pathology, Medical Genetics & Genomics, Uppsala University, Uppsala, Sweden (C.S.)
- Center for Computational Biology & Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India (J.K.)
- Framingham Heart Study (C.S.)
- Population Sciences Branch, National Heart, Lung & Blood Institute, National Institute of Health, Framingham, MA (C.S.)
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA (J.A.H.)
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, Sweden (N.L.P.)
- National Institute for Health and Welfare, Helsinki, Finland (M.P., V.S.)
- German Center for Diabetes Research (DZD), München-Neuherberg (C.G., A.P., H.G.)
- Clinical Cooperation Group Type 2 Diabetes (C.G., H.G.), Helmholtz Zentrum München, Neuherberg, Germany
- German Research Center for Environmental Health & Institute of Genetic Epidemiology (C.G., A.P.), Helmholtz Zentrum München, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology (H.G.), Helmholtz Zentrum München, Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics & Type 2 Diabetes (H.G.), Helmholtz Zentrum München, Neuherberg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (A.P.)
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands (S.M.W., C.v.D.)
- The Usher Institute of Population Health Sciences & Informatics (A.D.M.), University of Edinburgh, Edinburgh, U.K
- MRC Institute of Genetics & Molecular Medicine (H.M.C.), University of Edinburgh, Edinburgh, U.K
- Health Data Research UK, London, United Kingdom (A.D.M.)
- Department of Nutrition (Y.Z., F.B.H., L.Q.)
- Department of Epidemiology, Harvard School of Public Health, Boston, MA (F.B.H.)
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China (Y.Z.)
- University “Magna Graecia” of Catanzaro, Italy (G.S.)
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital & Harvard Medical School, Boston, MA (F.B.H.)
- Department of Epidemiology, School of Public Health & Tropical Medicine, Tulane University, New Orleans, LA (L.Q.)
- Faculty of Health Sciences, Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland (M.L.)
- Kuopio University Hospital, Finland (M.L.)
- Faculty of Medicine, University of Iceland. deCODE Genetics, Reykjavik, Iceland (U.T.)
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine (E.I.)
- Stanford Cardiovascular Institute (E.I.)
- Stanford Diabetes Research Center, Stanford University, Stanford, CA (E.I.)
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia (P.D.)
- Broad Institute of MIT & Harvard, Cambridge (S.K.)
- Cardiology Division, Center for Human Genetic Research (S.K.), Massachusetts General Hospital & Harvard Medical School, Boston, MA
- Cardiovascular Research Center (S.K.), Massachusetts General Hospital & Harvard Medical School, Boston, MA
- Heart & Lung Center, Helsinki University Hospital (J.S.) and Institute for Molecular Medicine Finland (FIMM), Helsinki University, Helsinki, Finland
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany (W.M.)
- Clinical Institute of Medical & Chemical Laboratory Diagnostics, Medical University of Graz, Austria (W.M.)
- Lerner Research Institute, Heart & Vascular Institute, Cleveland Clinic, Cleveland, OH (S.L.H.)
- Department of Biostatistics & Epidemiology, University of Pennsylvania, Philadelphia, PA (D.S.)
- Center for Non-Communicable Diseases, Karachi, Pakistan (D.S.)
- Department of Biostatistics, University of Liverpool, Liverpool, U.K. (A.P.M.)
- Division of Musculoskeletal & Dermatological Sciences, University of Manchester, Manchester, U.K. (A.P.M.)
- Public Health, NHS Fife, Kirkcaldy, Fife, U.K. (H.M.C.)
- Oxford NIHR Biomedical Research Center, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom (M.I.Mc)
| | - Christopher P. Nelson
- Department of Cardiovascular Sciences, University of Leicester (C.P.N., N.J.S.)
- NIHR Leicester Biomedical Research Center, Glenfield Hospital, Leicester, United Kingdom (C.P.N., N.J.S.)
| | - Lydia Coulter Kwee
- Duke Molecular Physiology Institute, Duke University, Durham, NC (L.C.K., S.H.S.)
| | - Tõnu Esko
- Estonian Genome Center (T.E., E.M., R.M., L.M., K.F., M.P., A. Metspalu), University of Tartu, Tartu, Estonia
| | - Evelin Mihailov
- Estonian Genome Center (T.E., E.M., R.M., L.M., K.F., M.P., A. Metspalu), University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center (T.E., E.M., R.M., L.M., K.F., M.P., A. Metspalu), University of Tartu, Tartu, Estonia
| | - Lili Milani
- Estonian Genome Center (T.E., E.M., R.M., L.M., K.F., M.P., A. Metspalu), University of Tartu, Tartu, Estonia
| | - Krista Fischer
- Estonian Genome Center (T.E., E.M., R.M., L.M., K.F., M.P., A. Metspalu), University of Tartu, Tartu, Estonia
| | - Stavroula Kanoni
- Center for Genomic Health (S.K.), Queen Mary University of London, London, United Kingdom
- William Harvey Research Institute, Barts & the London Medical School (S.K., P.D.), Queen Mary University of London, London, United Kingdom
- Broad Institute of MIT & Harvard, Cambridge (S.K.)
- Cardiology Division, Center for Human Genetic Research (S.K.), Massachusetts General Hospital & Harvard Medical School, Boston, MA
- Cardiovascular Research Center (S.K.), Massachusetts General Hospital & Harvard Medical School, Boston, MA
| | - Jitender Kumar
- Department of Medical Sciences, Molecular Epidemiology & Science for Life Laboratory (J.K., C.S., E.I.)
- Center for Computational Biology & Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India (J.K.)
| | - Ci Song
- Department of Medical Sciences, Molecular Epidemiology & Science for Life Laboratory (J.K., C.S., E.I.)
- Department of Immunology, Genetics and Pathology, Medical Genetics & Genomics, Uppsala University, Uppsala, Sweden (C.S.)
- Framingham Heart Study (C.S.)
- Population Sciences Branch, National Heart, Lung & Blood Institute, National Institute of Health, Framingham, MA (C.S.)
| | - Jaana A. Hartiala
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA (J.A.H.)
| | - Nancy L. Pedersen
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, Sweden (N.L.P.)
| | - Markus Perola
- Research Program for Clinical & Molecular Metabolism, Faculty of Medicine (M.P.), University of Helsinki, Helsinki, Finland. Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Estonian Genome Center (T.E., E.M., R.M., L.M., K.F., M.P., A. Metspalu), University of Tartu, Tartu, Estonia
- National Institute for Health and Welfare, Helsinki, Finland (M.P., V.S.)
| | - Christian Gieger
- German Center for Diabetes Research (DZD), München-Neuherberg (C.G., A.P., H.G.)
- Clinical Cooperation Group Type 2 Diabetes (C.G., H.G.), Helmholtz Zentrum München, Neuherberg, Germany
- German Research Center for Environmental Health & Institute of Genetic Epidemiology (C.G., A.P.), Helmholtz Zentrum München, Neuherberg, Germany
| | - Annette Peters
- German Center for Diabetes Research (DZD), München-Neuherberg (C.G., A.P., H.G.)
- German Research Center for Environmental Health & Institute of Genetic Epidemiology (C.G., A.P.), Helmholtz Zentrum München, Neuherberg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (A.P.)
| | - Liming Qu
- Cardiovascular Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA (L.Q., M.P.R.)
- Department of Nutrition (Y.Z., F.B.H., L.Q.)
- Department of Epidemiology, School of Public Health & Tropical Medicine, Tulane University, New Orleans, LA (L.Q.)
| | - Sara M. Willems
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands (S.M.W., C.v.D.)
| | - Alex S.F. Doney
- Division of Molecular & Clinical Medicine (A.S.F.D.), School of Medicine, University of Dundee
| | - Andrew D. Morris
- The Usher Institute of Population Health Sciences & Informatics (A.D.M.), University of Edinburgh, Edinburgh, U.K
- Health Data Research UK, London, United Kingdom (A.D.M.)
| | - Yan Zheng
- Department of Nutrition (Y.Z., F.B.H., L.Q.)
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China (Y.Z.)
| | - Giorgio Sesti
- University “Magna Graecia” of Catanzaro, Italy (G.S.)
| | - Frank B. Hu
- Department of Nutrition (Y.Z., F.B.H., L.Q.)
- Department of Epidemiology, Harvard School of Public Health, Boston, MA (F.B.H.)
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital & Harvard Medical School, Boston, MA (F.B.H.)
| | - Lu Qi
- Cardiovascular Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA (L.Q., M.P.R.)
- Department of Nutrition (Y.Z., F.B.H., L.Q.)
- Department of Epidemiology, School of Public Health & Tropical Medicine, Tulane University, New Orleans, LA (L.Q.)
| | - Markku Laakso
- Faculty of Health Sciences, Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland (M.L.)
- Kuopio University Hospital, Finland (M.L.)
| | | | - Harald Grallert
- German Center for Diabetes Research (DZD), München-Neuherberg (C.G., A.P., H.G.)
- Clinical Cooperation Group Type 2 Diabetes (C.G., H.G.), Helmholtz Zentrum München, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology (H.G.), Helmholtz Zentrum München, Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics & Type 2 Diabetes (H.G.), Helmholtz Zentrum München, Neuherberg, Germany
| | - Cornelia van Duijn
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands (S.M.W., C.v.D.)
| | - Muredach P. Reilly
- Cardiovascular Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA (L.Q., M.P.R.)
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology & Science for Life Laboratory (J.K., C.S., E.I.)
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine (E.I.)
- Stanford Cardiovascular Institute (E.I.)
- Stanford Diabetes Research Center, Stanford University, Stanford, CA (E.I.)
| | - Panos Deloukas
- William Harvey Research Institute, Barts & the London Medical School (S.K., P.D.), Queen Mary University of London, London, United Kingdom
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia (P.D.)
| | - Sek Kathiresan
- Center for Genomic Health (S.K.), Queen Mary University of London, London, United Kingdom
- William Harvey Research Institute, Barts & the London Medical School (S.K., P.D.), Queen Mary University of London, London, United Kingdom
- Broad Institute of MIT & Harvard, Cambridge (S.K.)
- Cardiology Division, Center for Human Genetic Research (S.K.), Massachusetts General Hospital & Harvard Medical School, Boston, MA
- Cardiovascular Research Center (S.K.), Massachusetts General Hospital & Harvard Medical School, Boston, MA
| | - Andres Metspalu
- Estonian Genome Center (T.E., E.M., R.M., L.M., K.F., M.P., A. Metspalu), University of Tartu, Tartu, Estonia
- Institute of Cell & Molecular Biology (A. Metspalu), University of Tartu, Tartu, Estonia
| | - Svati H. Shah
- Division of Cardiology, Department of Medicine, Duke University Medical Center (S.H.S.)
- Duke Molecular Physiology Institute, Duke University, Durham, NC (L.C.K., S.H.S.)
| | - Juha Sinisalo
- Heart & Lung Center, Helsinki University Hospital (J.S.) and Institute for Molecular Medicine Finland (FIMM), Helsinki University, Helsinki, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland (M.P., V.S.)
| | - Anders Hamsten
- Cardiovascular Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden (R.J.S., A.H.)
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester (C.P.N., N.J.S.)
- NIHR Leicester Biomedical Research Center, Glenfield Hospital, Leicester, United Kingdom (C.P.N., N.J.S.)
| | - Winfried März
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany (W.M.)
- Clinical Institute of Medical & Chemical Laboratory Diagnostics, Medical University of Graz, Austria (W.M.)
| | - Stanley L. Hazen
- Lerner Research Institute, Heart & Vascular Institute, Cleveland Clinic, Cleveland, OH (S.L.H.)
| | - Hugh Watkins
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
- Division of Cardiovascular Medicine (A.G., H.W.), University of Oxford, United Kingdom
| | - Danish Saleheen
- Department of Biostatistics & Epidemiology, University of Pennsylvania, Philadelphia, PA (D.S.)
- Center for Non-Communicable Diseases, Karachi, Pakistan (D.S.)
| | - Andrew P. Morris
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
- Department of Biostatistics, University of Liverpool, Liverpool, U.K. (A.P.M.)
- Division of Musculoskeletal & Dermatological Sciences, University of Manchester, Manchester, U.K. (A.P.M.)
| | - Helen M. Colhoun
- MRC Institute of Genetics & Molecular Medicine (H.M.C.), University of Edinburgh, Edinburgh, U.K
- Public Health, NHS Fife, Kirkcaldy, Fife, U.K. (H.M.C.)
| | - Leif Groop
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University Diabetes Center, Malmö, Sweden (C.L., L.G.)
| | - Mark I. McCarthy
- Oxford Center for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine (N.R.v.Z., N.W.R., N.R.R., A. Mahajan, M.I.Mc), University of Oxford, United Kingdom
- Wellcome Center for Human Genetics (N.R.v.Z., J.F.T., N.W.R., N.R.R, A. Mahajan, A.G., H.W., A.P.M., M.I.Mc), University of Oxford, United Kingdom
- Oxford NIHR Biomedical Research Center, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom (M.I.Mc)
| | - Colin N.A. Palmer
- Pat Macpherson Center for Pharmacogenetics & Pharmacogenomics, Cardiovascular & Diabetes Medicine (N.R.v.Z., C.N.A.P.), School of Medicine, University of Dundee
| |
Collapse
|
305
|
Gallo JE, Ochoa JE, Warren HR, Misas E, Correa MM, Gallo-Villegas JA, Bedoya G, Aristizábal D, McEwen JG, Caulfield MJ, Parati G, Clay OK. Hypertension and the roles of the 9p21.3 risk locus: Classic findings and new association data. Int J Cardiol Hypertens 2020; 7:100050. [PMID: 33330845 PMCID: PMC7491459 DOI: 10.1016/j.ijchy.2020.100050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The band 9p21.3 contains an established genomic risk zone for cardiovascular disease (CVD). Since the initial 2007 Wellcome Trust Case Control Consortium study (WTCCC), the increased CVD risk associated with 9p21.3 has been confirmed by multiple studies in different continents. However, many years later there was still no confirmed report of a corresponding association of 9p21.3 with hypertension, a major CV risk factor, nor with blood pressure (BP). THEORY In this contribution, we review the bipartite haplotype structure of the 9p21.3 risk locus: one block is devoid of protein-coding genes but contains the lead CVD risk SNPs, while the other block contains the first exon and regulatory DNA of the gene for the cell cycle inhibitor p15. We consider how findings from molecular biology offer possibilities of an involvement of p15 in hypertension etiology, with expression of the p15 gene modulated by genetic variation from within the 9p21.3 risk locus. RESULTS We present original results from a Colombian study revealing moderate but persistent association signals for BP and hypertension within the classic 9p21.3 CVD risk locus. These SNPs are mostly confined to a 'hypertension island' that spans less than 60 kb and coincides with the p15 haplotype block. We find confirmation in data originating from much larger, recent European BP studies, albeit with opposite effect directions. CONCLUSION Although more work will be needed to elucidate possible mechanisms, previous findings and new data prompt reconsidering the question of how variation in 9p21.3 might influence hypertension components of cardiovascular risk.
Collapse
Key Words
- 1 KG, 1000 Genomes Project
- BP, blood pressure
- Blood pressure levels
- CVD, cardiovascular disease
- DBP, diastolic blood pressure
- EGFR, epidermal growth factor receptor
- GWAS, genome wide association studi(es)
- Genotype-phenotype associations
- Haplotypes
- MAF, minor allele frequency
- RAS, renin angiotensin system
- SBP, systolic blood pressure
- SNP, single nucleotide polymorphism
- TGF-β, transforming growth factor beta
- VSMC, vascular smooth muscle cell(s)
- bp, base pair
- kb, kilobase pair
Collapse
Affiliation(s)
- Juan E. Gallo
- Cellular & Molecular Biology Unit, Corporación para Investigaciones Biológicas, Medellín, Colombia
- Doctoral Program in Biomedical Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Juan E. Ochoa
- Cellular & Molecular Biology Unit, Corporación para Investigaciones Biológicas, Medellín, Colombia
- Istituto Auxologico Italiano, IRCCS, Department of Cardiovascular Neural and Metabolic Sciences, San Luca Hospital, Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Helen R. Warren
- Clinical Pharmacology Department, 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 Center, Queen Mary University of London, London, UK
| | - Elizabeth Misas
- Cellular & Molecular Biology Unit, Corporación para Investigaciones Biológicas, Medellín, Colombia
- Institute of Biology, Universidad de Antioquia, Medellín, Colombia
| | | | | | - Gabriel Bedoya
- Institute of Biology, Universidad de Antioquia, Medellín, Colombia
| | - Dagnóvar Aristizábal
- Cellular & Molecular Biology Unit, Corporación para Investigaciones Biológicas, Medellín, Colombia
- SICOR, Medellín, Colombia
| | - Juan G. McEwen
- Cellular & Molecular Biology Unit, Corporación para Investigaciones Biológicas, Medellín, Colombia
- School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Mark J. Caulfield
- Clinical Pharmacology Department, 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 Center, Queen Mary University of London, London, UK
| | - Gianfranco Parati
- Istituto Auxologico Italiano, IRCCS, Department of Cardiovascular Neural and Metabolic Sciences, San Luca Hospital, Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Oliver K. Clay
- Cellular & Molecular Biology Unit, Corporación para Investigaciones Biológicas, Medellín, Colombia
- Translational Microbiology and Emerging Diseases (MICROS), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
306
|
Wu T, Wang Y, Shi W, Zhang BQ, Raelson J, Yao YM, Wu HD, Xu ZX, Marois-Blanchet FC, Ledoux J, Blunck R, Sheng JZ, Hu SJ, Luo H, Wu J. A Variant in the Nicotinic Acetylcholine Receptor Alpha 3 Subunit Gene Is Associated With Hypertension Risks in Hypogonadic Patients. Front Genet 2020; 11:539862. [PMID: 33329690 PMCID: PMC7728919 DOI: 10.3389/fgene.2020.539862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 11/10/2020] [Indexed: 12/29/2022] Open
Abstract
Ephb6 gene knockout causes hypertension in castrated mice. EPHB6 controls catecholamine secretion by adrenal gland chromaffin cells (AGCCs) in a testosterone-dependent way. Nicotinic acetylcholine receptor (nAChR) is a ligand-gated Ca2+/Na+ channel, and its opening is the first signaling event leading to catecholamine secretion by AGCCs. There is a possibility that nAChR might be involved in EPHB6 signaling, and thus sequence variants of its subunit genes are associated with hypertension risks. CHRNA3 is the major subunit of nAChR used in human and mouse AGCCs. We conducted a human genetic study to assess the association of CHRNA3 variants with hypertension risks in hypogonadic males. The study cohort included 1,500 hypogonadic Chinese males with (750 patients) or without (750 patients) hypertension. The result revealed that SNV rs3743076 in the fourth intron of CHRNA3 was significantly associated with hypertension risks in the hypogonadic males. We further showed that EPHB6 physically interacted with CHRNA3 in AGCCs, providing a molecular basis for nAChR being in the EPHB6 signaling pathway.
Collapse
Affiliation(s)
- Tao Wu
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yujia Wang
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.,Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Shi
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Bi-Qi Zhang
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - John Raelson
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Yu-Mei Yao
- Department of Cardiology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Huan-Dong Wu
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zao-Xian Xu
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | | | - Jonathan Ledoux
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Rikard Blunck
- Department of Physics, University of Montreal, Montreal, QC, Canada
| | - Jian-Zhong Sheng
- Department of Pathology and Physiopathology, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shen-Jiang Hu
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hongyu Luo
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Jiangping Wu
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.,Nephrology Service, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| |
Collapse
|
307
|
Teleka S, Hindy G, Drake I, Poveda A, Melander O, Liedberg F, Orho-Melander M, Stocks T. Blood pressure and bladder cancer risk in men by use of survival analysis and in interaction with NAT2 genotype, and by Mendelian randomization analysis. PLoS One 2020; 15:e0241711. [PMID: 33237904 PMCID: PMC7688142 DOI: 10.1371/journal.pone.0241711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022] Open
Abstract
The association between blood pressure (BP) and bladder cancer (BC) risk remains unclear with confounding by smoking being of particular concern. We investigated the association between BP and BC risk among men using conventional survival-analysis, and by Mendelian Randomization (MR) analysis in an attempt to disconnect the association from smoking. We additionally investigated the interaction between BP and N-acetyltransferase-2 (NAT2) rs1495741, an established BC genetic risk variant, in the association. Populations consisting of 188,167 men with 502 incident BC's in the UK-biobank and 27,107 men with 928 incident BC's in two Swedish cohorts were used for the analysis. We found a positive association between systolic BP and BC risk in Cox-regression survival analysis in the Swedish cohorts, (hazard ratio [HR] per standard deviation [SD]: 1.14 [95% confidence interval 1.05-1.22]) and MR analysis (odds ratio per SD: 2-stage least-square regression, 7.70 [1.92-30.9]; inverse-variance weighted estimate, 3.43 [1.12-10.5]), and no associations in the UK-biobank (HR systolic BP: 0.93 [0.85-1.02]; MR OR: 1.24 [0.35-4.40] and 1.37 [0.43-4.37], respectively). BP levels were positively associated with muscle-invasive BC (MIBC) (HRs: systolic BP, 1.32 [1.09-1.59]; diastolic BP, 1.27 [1.04-1.55]), but not with non-muscle invasive BC, which could be analyzed in the Swedish cohorts only. There was no interaction between BP and NAT2 in relation to BC on the additive or multiplicative scale. These results suggest that BP might be related to BC, more particularly MIBC. There was no evidence to support interaction between BP and NAT2 in relation to BC in our study.
Collapse
Affiliation(s)
- Stanley Teleka
- Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
- * E-mail:
| | - George Hindy
- Department of Population Medicine, College of Medicine Qatar University, Doha, Qatar
- Broad Institute, Cambridge, Massachusetts, United States of America
| | - Isabel Drake
- Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Alaitz Poveda
- Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Olle Melander
- Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Fredrik Liedberg
- Division of Urological Research, Institution of Translational Medicine, Lund University, Malmö, Sweden
- Department of Urology, Skåne University Hospital, Skåne, Sweden
| | | | - Tanja Stocks
- Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
| |
Collapse
|
308
|
Kolbeinsson A, Filippi S, Panagakis Y, Matthews PM, Elliott P, Dehghan A, Tzoulaki I. Accelerated MRI-predicted brain ageing and its associations with cardiometabolic and brain disorders. Sci Rep 2020; 10:19940. [PMID: 33203906 PMCID: PMC7672070 DOI: 10.1038/s41598-020-76518-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Brain structure in later life reflects both influences of intrinsic aging and those of lifestyle, environment and disease. We developed a deep neural network model trained on brain MRI scans of healthy people to predict "healthy" brain age. Brain regions most informative for the prediction included the cerebellum, hippocampus, amygdala and insular cortex. We then applied this model to data from an independent group of people not stratified for health. A phenome-wide association analysis of over 1,410 traits in the UK Biobank with differences between the predicted and chronological ages for the second group identified significant associations with over 40 traits including diseases (e.g., type I and type II diabetes), disease risk factors (e.g., increased diastolic blood pressure and body mass index), and poorer cognitive function. These observations highlight relationships between brain and systemic health and have implications for understanding contributions of the latter to late life dementia risk.
Collapse
Affiliation(s)
- Arinbjörn Kolbeinsson
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK.
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, W2 1PG, UK.
| | - Sarah Filippi
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, W2 1PG, UK
- Department of Mathematics, Imperial College London, London, SW7 2AZ, UK
| | - Yannis Panagakis
- Department of Computing, Imperial College London, London, SW7 2AZ, UK
- Department of Informatics and Telecommunications, University of Athens, Athens, Greece
| | - Paul M Matthews
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, London, W12 0NN, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, W2 1PG, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
- National Institute for Health Research, Imperial Biomedical Research Centre, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
- Health Data Research UK London at Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, W2 1PG, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, W2 1PG, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| |
Collapse
|
309
|
Diet-gene interaction: effects of polymorphisms in the ACE, AGT and BDKRB2 genes and the consumption of sodium, potassium, calcium, and magnesium on blood pressure of normotensive adult individuals. Mol Cell Biochem 2020; 476:1211-1219. [PMID: 33190196 DOI: 10.1007/s11010-020-03983-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
Functional variants in genes of the renin-angiotensin (RAS) and kallikrein-kinin (KKS) systems have already been implicated in blood pressure (BP) modulation, but few studies have focused on a nutrigenetics approach. Thus, the aim of this study is to verify the effects of the interaction between genetic polymorphisms (rs4340-ACE, rs699-AGT, and rs1799722-BDKRB2) and micronutrient consumption (sodium, potassium, calcium, and magnesium) on BP values of normotensive adult individuals. The study included 335 adults, men and women, 25.5 (6.6) years old. Biochemical, anthropometric, BP measurements, and food intake data were assessed for all participants. Gene-nutrient interaction on BP outcome was tested by multiple linear regression with manual backward stepwise modeling. Our results indicated that individuals with G allele for rs699 polymorphism, in the increase of sodium and magnesium consumption, both in the genotypic model (sodium, p = 0.035; magnesium, p = 0.016) and in the dominant model (sodium, p = 0.009; magnesium, p = 0.006) had higher systolic BP (SBP) levels compared to AA homozygotes (sodium, p = 0.001; magnesium, p < 0.001). Also, individuals with the T allele for the rs1799722 polymorphism, with higher calcium intake, had significantly higher levels of SBP and diastolic BP (DBP) when compared to CC homozygotes (p = 0.037). In conclusion, our findings pointed for significant interactions between genetic polymorphisms (rs699-AGT and rs1799722-BDKRB2) and the consumption of micronutrients (sodium, magnesium, and calcium) on the BP variation. These findings contribute to the understanding of the complex mechanisms involved in BP regulation, which probable include several gene-nutrition interactions.
Collapse
|
310
|
Dong X, Zhu Z, Wei Y, Ngo D, Zhang R, Du M, Huang H, Lin L, Tejera P, Su L, Chen F, Ahasic AM, Thompson BT, Meyer NJ, Christiani DC. Plasma Insulin-like Growth Factor Binding Protein 7 Contributes Causally to ARDS 28-Day Mortality: Evidence From Multistage Mendelian Randomization. Chest 2020; 159:1007-1018. [PMID: 33189655 DOI: 10.1016/j.chest.2020.10.074] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND ARDS is a devastating syndrome with heterogeneous subtypes, but few causal biomarkers have been identified. RESEARCH QUESTION Would multistage Mendelian randomization identify new causal protein biomarkers for ARDS 28-day mortality? STUDY DESIGN AND METHODS Three hundred moderate to severe ARDS patients were selected randomly from the Molecular Epidemiology of ARDS cohort for proteomics analysis. Orthogonal projections to latent structures discriminant analysis was applied to detect the association between proteins and ARDS 28-day mortality. Candidate proteins were analyzed using generalized summary data-based Mendelian randomization (GSMR). Protein quantitative trait summary statistics were retrieved from the Efficiency and safety of varying the frequency of whole blood donation (INTERVAL) study (n = 2,504), and a genome-wide association study for ARDS was conducted from the Identification of SNPs Predisposing to Altered Acute Lung Injury Risk (iSPAAR) consortium study (n = 534). Causal mediation analysis detected the role of platelet count in mediating the effect of protein on ARDS prognosis. RESULTS Plasma insulin-like growth factor binding protein 7 (IGFBP7) moderately increased ARDS 28-day mortality (OR, 1.11; 95% CI, 1.04-1.19; P = .002) per log2 increase. GSMR analysis coupled with four other Mendelian randomization methods revealed IGFBP7 as a causal biomarker for ARDS 28-day mortality (OR, 2.61; 95% CI, 1.33-5.13; P = .005). Causal mediation analysis indicated that the association between IGFBP7 and ARDS 28-day mortality is mediated by platelet count (OR, 1.03; 95% CI, 1.02-1.04; P = .01). INTERPRETATION We identified plasma IGFBP7 as a novel causal protein involved in the pathogenesis of ARDS 28-day mortality and platelet function in ARDS, a topic for further experimental and clinical investigation.
Collapse
Affiliation(s)
- Xuesi Dong
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Zhaozhong Zhu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Yongyue Wei
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Debby Ngo
- Pulmonary, Critical Care & Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Ruyang Zhang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mulong Du
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Huang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lijuan Lin
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Paula Tejera
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Feng Chen
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Amy M Ahasic
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA; Section of Pulmonary and Critical Care Medicine, Norwalk Hospital, Nuvance Health, Norwalk, CT
| | - B Taylor Thompson
- Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Nuala J Meyer
- Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA.
| |
Collapse
|
311
|
Ward M, Hughes CF, Strain JJ, Reilly R, Cunningham C, Molloy AM, Horigan G, Casey M, McCarroll K, O'Kane M, Gibney MJ, Flynn A, Walton J, McNulty BA, McCann A, Kirwan L, Scott JM, McNulty H. Impact of the common MTHFR 677C→T polymorphism on blood pressure in adulthood and role of riboflavin in modifying the genetic risk of hypertension: evidence from the JINGO project. BMC Med 2020; 18:318. [PMID: 33172445 PMCID: PMC7656675 DOI: 10.1186/s12916-020-01780-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/10/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Genome-wide and clinical studies have linked the 677C→T polymorphism in the gene encoding methylenetetrahydrofolate reductase (MTHFR) with hypertension, whilst limited evidence shows that intervention with riboflavin (i.e. the MTHFR co-factor) can lower blood pressure (BP) in hypertensive patients with the variant MTHFR 677TT genotype. We investigated the impact of this common polymorphism on BP throughout adulthood and hypothesised that riboflavin status would modulate the genetic risk of hypertension. METHODS Observational data on 6076 adults of 18-102 years were drawn from the Joint Irish Nutrigenomics Organisation project, comprising the Trinity-Ulster Department of Agriculture (TUDA; volunteer sample) and the National Adult Nutrition Survey (NANS; population-based sample) cohorts. Participants were recruited from the Republic of Ireland and Northern Ireland (UK) in 2008-2012 using standardised methods. RESULTS The variant MTHFR 677TT genotype was identified in 12% of adults. From 18 to 70 years, this genotype was associated with an increased risk of hypertension (i.e. systolic BP ≥ 140 and/or a diastolic BP ≥ 90 mmHg): odds ratio (OR) 1.42, 95% confidence interval (CI) 1.07 to 1.90; P = 0.016, after adjustment for antihypertensive drug use and other significant factors, namely, age, male sex, BMI, alcohol and total cholesterol. Low or deficient biomarker status of riboflavin (observed in 30.2% and 30.0% of participants, respectively) exacerbated the genetic risk of hypertension, with a 3-fold increased risk for the TT genotype in combination with deficient riboflavin status (OR 3.00, 95% CI, 1.34-6.68; P = 0.007) relative to the CC genotype combined with normal riboflavin status. Up to 65 years, we observed poorer BP control rates on antihypertensive treatment in participants with the TT genotype (30%) compared to those without this variant, CT (37%) and CC (45%) genotypes (P < 0.027). CONCLUSIONS The MTHFR 677TT genotype is associated with higher BP independently of homocysteine and predisposes adults to an increased risk of hypertension and poorer BP control with antihypertensive treatment, whilst better riboflavin status is associated with a reduced genetic risk. Riboflavin intervention may thus offer a personalised approach to prevent the onset of hypertension in adults with the TT genotype; however, this requires confirmation in a randomised trial in non-hypertensive adults.
Collapse
Affiliation(s)
- Mary Ward
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Catherine F Hughes
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - J J Strain
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Rosie Reilly
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Conal Cunningham
- The Department of Gerontology, St James's Hospital, Dublin, Ireland
| | - Anne M Molloy
- School of Medicine and School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Geraldine Horigan
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Miriam Casey
- The Department of Gerontology, St James's Hospital, Dublin, Ireland
| | - Kevin McCarroll
- The Department of Gerontology, St James's Hospital, Dublin, Ireland
| | - Maurice O'Kane
- Clinical Chemistry Laboratory, Western Health and Social Care Trust, Altnagelvin Hospital, Londonderry, Northern Ireland, UK
| | - Michael J Gibney
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Albert Flynn
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - Janette Walton
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - Breige A McNulty
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Adrian McCann
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Laura Kirwan
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - John M Scott
- School of Medicine and School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Helene McNulty
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| |
Collapse
|
312
|
Wang H, Liu Z, Shao J, Jiang M, Lu X, Lin L, Wang L, Xu Q, Zhang H, Li X, Zhou J, Chen Y, Zhang R. Pathogenesis of premature coronary artery disease: Focus on risk factors and genetic variants. Genes Dis 2020; 9:370-380. [PMID: 35224153 PMCID: PMC8843894 DOI: 10.1016/j.gendis.2020.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/17/2020] [Accepted: 11/04/2020] [Indexed: 11/24/2022] Open
Abstract
The development of premature coronary artery disease (PCAD) is dependent on both genetic predisposition and traditional risk factors. Strategies for unraveling the genetic basis of PCAD have evolved with the advent of modern technologies. Genome-wide association studies (GWASs) have identified a considerable number of common genetic variants that are associated with PCAD. Most of these genetic variants are attributable to lipid and blood pressure-related single-nucleotide polymorphisms (SNPs). The genetic variants that predispose individuals to developing PCAD may depend on race and ethnicity. Some characteristic genetic variants have been identified in Chinese populations. Although translating this genetic knowledge into clinical applications is still challenging, these genetic variants can be used for CAD phenotype identification, genetic prediction and therapy. In this article we will provide a comprehensive review of genetic variants detected by GWASs that are predicted to contribute to the development of PCAD. We will highlight recent findings regarding CAD-related genetic variants in Chinese populations and discuss the potential clinical utility of genetic variants for preventing and managing PCAD.
Collapse
|
313
|
Atkins JL, Masoli JAH, Delgado J, Pilling LC, Kuo CL, Kuchel GA, Melzer D. Preexisting Comorbidities Predicting COVID-19 and Mortality in the UK Biobank Community Cohort. J Gerontol A Biol Sci Med Sci 2020; 75:2224-2230. [PMID: 32687551 PMCID: PMC7454409 DOI: 10.1093/gerona/glaa183] [Citation(s) in RCA: 320] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Hospitalized COVID-19 patients tend to be older and frequently have hypertension, diabetes, or coronary heart disease, but whether these comorbidities are true risk factors (ie, more common than in the general older population) is unclear. We estimated associations between preexisting diagnoses and hospitalized COVID-19 alone or with mortality, in a large community cohort. METHODS UK Biobank (England) participants with baseline assessment 2006-2010, followed in hospital discharge records to 2017 and death records to 2020. Demographic and preexisting common diagnoses association tested with hospitalized laboratory-confirmed COVID-19 (March 16 to April 26, 2020), alone or with mortality, in logistic models. RESULTS Of 269 070 participants aged older than 65, 507 (0.2%) became COVID-19 hospital inpatients, of which 141 (27.8%) died. Common comorbidities in hospitalized inpatients were hypertension (59.6%), history of fall or fragility fractures (29.4%), coronary heart disease (21.5%), type 2 diabetes (type 2, 19. 9%), and asthma (17.6%). However, in models adjusted for comorbidities, age group, sex, ethnicity, and education, preexisting diagnoses of dementia, type 2 diabetes, chronic obstructive pulmonary disease, pneumonia, depression, atrial fibrillation, and hypertension emerged as independent risk factors for COVID-19 hospitalization, the first 5 remaining statistically significant for related mortality. Chronic kidney disease and asthma were risk factors for COVID-19 hospitalization in women but not men. CONCLUSIONS There are specific high-risk preexisting comorbidities for COVID-19 hospitalization and related deaths in community-based older men and women. These results do not support simple age-based targeting of the older population to prevent severe COVID-19 infections.
Collapse
Affiliation(s)
- Janice L Atkins
- Epidemiology and Public Health Group, University of Exeter Medical School, UK
| | - Jane A H Masoli
- Epidemiology and Public Health Group, University of Exeter Medical School, UK.,Department of Healthcare for Older People, Royal Devon and Exeter Hospital, UK
| | - Joao Delgado
- Epidemiology and Public Health Group, University of Exeter Medical School, UK
| | - Luke C Pilling
- Epidemiology and Public Health Group, University of Exeter Medical School, UK.,Center on Aging, University of Connecticut Health Center, Farmington
| | - Chia-Ling Kuo
- Center on Aging, University of Connecticut Health Center, Farmington
| | - George A Kuchel
- Center on Aging, University of Connecticut Health Center, Farmington
| | - David Melzer
- Epidemiology and Public Health Group, University of Exeter Medical School, UK.,Center on Aging, University of Connecticut Health Center, Farmington
| |
Collapse
|
314
|
Sun YV, Liu C, Staimez L, Ali MK, Chang H, Kondal D, Patel S, Jones D, Mohan V, Tandon N, Prabhakaran D, Quyyumi AA, Narayan KMV, Agrawal A. Cardiovascular disease risk and pathophysiology in South Asians: can longitudinal multi-omics shed light? Wellcome Open Res 2020; 5:255. [PMID: 34136649 PMCID: PMC8176264 DOI: 10.12688/wellcomeopenres.16336.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2020] [Indexed: 03/27/2025] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality in South Asia, with rapidly increasing prevalence of hypertension, type 2 diabetes and hyperlipidemia over the last two decades. Atherosclerotic CVD (ASCVD) affects South Asians earlier in life and at lower body weights, which is not fully explained by differential burden of conventional risk factors. Heart failure (HF) is a complex clinical syndrome of heterogeneous structural phenotypes including two major clinical subtypes, HF with preserved (HFpEF) and reduced ejection fraction (HFrEF). The prevalence of HF in South Asians is also rising with other metabolic diseases, and HFpEF develops at younger age and leaner body mass index in South Asians than in Whites. Recent genome-wide association studies, epigenome-wide association studies and metabolomic studies of ASCVD and HF have identified genes, metabolites and pathways associated with CVD traits. However, these findings were mostly driven by samples of European ancestry, which may not accurately represent the CVD risk at the molecular level, and the unique risk profile of CVD in South Asians. Such bias, while formulating hypothesis-driven research studies, risks missing important causal or predictive factors unique to South Asians. Importantly, a longitudinal design of multi-omic markers can capture the life-course risk and natural history related to CVD, and partially disentangle putative causal relationship between risk factors, multi-omic markers and subclinical and clinical ASCVD and HF. In conclusion, combining high-resolution untargeted metabolomics with epigenomics of rigorous, longitudinal design will provide comprehensive unbiased molecular characterization of subclinical and clinical CVD among South Asians. A thorough understanding of CVD-associated metabolomic profiles, together with advances in epigenomics and genomics, will lead to more accurate estimates of CVD progression and stimulate new strategies for improving cardiovascular health.
Collapse
Affiliation(s)
- Yan V. Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
- Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Chang Liu
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Lisa Staimez
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Mohammed K. Ali
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
- Department of Family and Preventive Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Howard Chang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | | | - Shivani Patel
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Dean Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | | | - Nikhil Tandon
- All India Institute of Medical Sciences, New Delhi, India
| | | | - Arshed A. Quyyumi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - K. M. Venkat Narayan
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Anurag Agrawal
- Institute of Genomics and Integrative Biology, Council of Scientific and Industrial Research, New Delhi, India
| |
Collapse
|
315
|
Ali W, Bakris GL. How to Manage Hypertension in People With Diabetes. Am J Hypertens 2020; 33:935-943. [PMID: 32307510 DOI: 10.1093/ajh/hpaa067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/13/2020] [Accepted: 04/15/2020] [Indexed: 11/14/2022] Open
Abstract
Hypertension is a common condition that is often seen in patients with diabetes. Both diseases increase the risk of morbidity and mortality from CV events and kidney disease progression. Factors that influence blood pressure (BP) control in diabetes include the persons' genetic background for hypertension and kidney disease, level of obesity and insulin resistance, the magnitude of preexisting kidney disease, and lifestyle factors, such as level of sodium and potassium intake, sleep quality and exercise effort all of which can affect levels of sympathetic nerve activity and contribute to increased BP variability. Lifestyle intervention is a key component to the effective management of diabetes and hypertension and can markedly reduce event rates of both heart and kidney outcomes. The approach to pharmacologic treatment of BP in diabetes is crucial since certain classes of agents for both BP and diabetes confer significant benefits to reduce cardiorenal outcomes.
Collapse
Affiliation(s)
- Waleed Ali
- Department of Medicine, American Heart Association Comprehensive Hypertension Center, Section of Endocrinology, Diabetes and Metabolism, The University of Chicago Medicine, Chicago, Illinois, USA
| | - George L Bakris
- Department of Medicine, American Heart Association Comprehensive Hypertension Center, Section of Endocrinology, Diabetes and Metabolism, The University of Chicago Medicine, Chicago, Illinois, USA
| |
Collapse
|
316
|
Laird E, O'halloran AM, Fedorowski A, Melander O, Hever A, Sjögren M, Carey D, Kenny RA. Orthostatic Hypotension and Novel Blood Pressure Associated Gene Variants in Older Adults: Data From the TILDA Study. J Gerontol A Biol Sci Med Sci 2020; 75:2074-2080. [PMID: 31821404 DOI: 10.1093/gerona/glz286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Indexed: 11/14/2022] Open
Abstract
Orthostatic hypotension (OH) is associated with increased risk of trauma and cardiovascular events. Recent studies have identified new genetic variants that influence orthostatic blood pressure (BP). The aim of this study was to investigate the associations of candidate gene loci with orthostatic BP responses in older adults. A total of 3,430 participants aged ≥50 years from The Irish Longitudinal Study on Ageing (TILDA) with BP measures and genetic data from 12 single-nucleotide polymorphism (SNP) linked to BP responses were analyzed. Orthostatic BP responses were recorded at each 10 s interval and were defined as OH (SBP drop ≥20 mmHg or DBP drop ≥10 mmHg) at the time-points 40, 90, and 110 s. We defined sustained OH (SOH) as a drop that exceeded consensus BP thresholds for OH at 40, 90, and 110 s after standing. Logistic regression analyses modeled associations between the candidate SNP alleles and OH. We report no significant associations between OH and measured SNPs after correction for multiple comparisons apart from the SNP rs5068 where proportion of the minor allele was significantly different between cases and controls for SOH 40 (p = .002). After adjustment for covariates in a logistic regression, those with the minor G allele (compared to the A allele) had a decreased incidence rate ratio (IRR) for SOH 40 (IRR 0.45, p = .001, 95% CI 0.29-0.72). Only one SNP linked with increased natriuretic peptide concentrations was associated with OH. These results suggest that genetic variants may have a weak impact on OH but needs verification in other population studies.
Collapse
Affiliation(s)
- Eamon Laird
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Ireland
| | | | - Artur Fedorowski
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden.,Department of Clinical Sciences, Clinical Research Center, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Clinical Research Center, Lund University, Malmö, Sweden
| | - Ann Hever
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Ireland
| | - Marketa Sjögren
- Department of Clinical Sciences, Clinical Research Center, Lund University, Malmö, Sweden
| | - Daniel Carey
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Ireland
| | - Rose Anne Kenny
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Ireland.,Mercer's Institute for Successful Ageing, St. James's Hospital, Dublin, Ireland
| |
Collapse
|
317
|
Morabito S, Miyoshi E, Michael N, Swarup V. Integrative genomics approach identifies conserved transcriptomic networks in Alzheimer's disease. Hum Mol Genet 2020; 29:2899-2919. [PMID: 32803238 PMCID: PMC7566321 DOI: 10.1093/hmg/ddaa182] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/10/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurological disorder characterized by changes in cell-type proportions and consequently marked alterations of the transcriptome. Here we use a data-driven systems biology meta-analytical approach across three human AD cohorts, encompassing six cortical brain regions, and integrate with multi-scale datasets comprising of DNA methylation, histone acetylation, transcriptome- and genome-wide association studies and quantitative trait loci to further characterize the genetic architecture of AD. We perform co-expression network analysis across more than 1200 human brain samples, identifying robust AD-associated dysregulation of the transcriptome, unaltered in normal human aging. We assess the cell-type specificity of AD gene co-expression changes and estimate cell-type proportion changes in human AD by integrating co-expression modules with single-cell transcriptome data generated from 27 321 nuclei from human postmortem prefrontal cortical tissue. We also show that genetic variants of AD are enriched in a microglial AD-associated module and identify key transcription factors regulating co-expressed modules. Additionally, we validate our results in multiple published human AD gene expression datasets, which can be easily accessed using our online resource (https://swaruplab.bio.uci.edu/consensusAD).
Collapse
Affiliation(s)
- Samuel Morabito
- Mathematical, Computational and Systems Biology (MCSB) Program, University of California, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697, USA
| | - Emily Miyoshi
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697, USA
| | - Neethu Michael
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697, USA
| |
Collapse
|
318
|
Kollarova M, Puzserova A, Balis P, Radosinska D, Tothova L, Bartekova M, Barancik M, Radosinska J. Age- and Phenotype-Dependent Changes in Circulating MMP-2 and MMP-9 Activities in Normotensive and Hypertensive Rats. Int J Mol Sci 2020; 21:E7286. [PMID: 33023122 PMCID: PMC7582756 DOI: 10.3390/ijms21197286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 12/27/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are important in the pathogenesis of numerous diseases. The present study aimed to monitor the activation of MMP-2 and MMP-9 in spontaneously hypertensive rats (SHR) and their normotensive counterparts-Wistar-Kyoto rats (WKY). The animals were divided according to age (7, 20, and 52 weeks) and phenotype into: WKY-7, WKY-20, WKY-52, SHR-7, SHR-20 and SHR-52 groups. MMP plasma activities were determined by gelatine zymography. We monitored selected parameters of oxidative stress and antioxidant status. N-terminal pro-brain natriuretic peptide (NT-proBNP) was determined as a marker of heart function and neurohumoral activation. SHR-7 showed higher MMP-2 activity compared with WKY-7, while SHR-52 showed lower MMP-2 and MMP-9 activities compared with WKY-52. Examining age-dependent changes in MMP activities, we found a decrease in MMP-2 activity and increase in MMP-9 activity with increasing age in both phenotypes. Parameters of oxidative stress and antioxidant status as well as NT-proBNP levels were not significantly worsened due to aging in SHR. Our results suggest that hypertension is accompanied by varying MMP activation during aging. The results of our study may indicate that MMP-2 inhibition is therapeutically applicable during the development of hypertension, while in developed, stabilized and uncomplicated hypertension, systemic MMP-2 and MMP-9 inhibition may not be desirable.
Collapse
Affiliation(s)
- Marta Kollarova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia; (M.K.); (M.B.)
| | - Angelika Puzserova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute of Normal and Pathological Physiology, 813 71 Bratislava, Slovakia; (A.P.); (P.B.)
| | - Peter Balis
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute of Normal and Pathological Physiology, 813 71 Bratislava, Slovakia; (A.P.); (P.B.)
| | - Dominika Radosinska
- Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia;
| | - Lubomira Tothova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia;
| | - Monika Bartekova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia; (M.K.); (M.B.)
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 841 04 Bratislava, Slovakia;
| | - Miroslav Barancik
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 841 04 Bratislava, Slovakia;
| | - Jana Radosinska
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia; (M.K.); (M.B.)
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 841 04 Bratislava, Slovakia;
| |
Collapse
|
319
|
Tegegne BS, Man T, van Roon AM, Asefa NG, Riese H, Nolte I, Snieder H. Heritability and the Genetic Correlation of Heart Rate Variability and Blood Pressure in >29 000 Families: The Lifelines Cohort Study. Hypertension 2020; 76:1256-1262. [PMID: 32829661 PMCID: PMC7480943 DOI: 10.1161/hypertensionaha.120.15227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/05/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022]
Abstract
Dysregulation of the cardiac autonomic nervous system, as indexed by reduced heart rate variability (HRV), has been associated with the development of high blood pressure (BP). However, the underlying pathological mechanisms are not yet fully understood. This study aimed to estimate heritability of HRV and BP and to determine their genetic overlap. We used baseline data of the 3-generation Lifelines population-based cohort study (n=149 067; mean age, 44.5). In-house software was used to calculate root mean square of successive differences and SD of normal-to-normal intervals as indices of HRV based on 10-second resting ECGs. BP was recorded with an automatic BP monitor. We estimated heritabilities and genetic correlations with variance components methods in ASReml software. We additionally estimated genetic correlations with bivariate linkage disequilibrium score regression using publicly available genome-wide association study data. The heritability (SE) estimates were 15.6% (0.90%) for SD of normal-to-normal intervals and 17.9% (0.90%) for root mean square of successive differences. For BP measures, they ranged from 24.4% (0.90%) for pulse pressure to 30.3% (0.90%) for diastolic BP. Significant negative genetic correlations (all P<0.0001) of root mean square of successive differences/SD of normal-to-normal intervals with systolic BP (-0.20/-0.16) and with diastolic BP (-0.15/-0.13) were observed. LD score regression showed largely consistent genetic correlation estimates of root mean square of successive differences/SD of normal-to-normal intervals with systolic BP (range, -0.08 to -0.23) and diastolic BP (range, -0.20 to -0.27). Our study shows a substantial contribution of genetic factors in explaining the variance of HRV and BP measures in the general population. The significant negative genetic correlations between HRV and BP indicate that genetic pathways for HRV and BP partially overlap.
Collapse
Affiliation(s)
- Balewgizie S. Tegegne
- From the Department of Epidemiology (B.S.T., T.M., N.G.A., I.N., H.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Tengfei Man
- From the Department of Epidemiology (B.S.T., T.M., N.G.A., I.N., H.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Arie M. van Roon
- Department of Vascular Medicine (A.M.v.R.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Nigus G. Asefa
- From the Department of Epidemiology (B.S.T., T.M., N.G.A., I.N., H.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Harriëtte Riese
- Department of Psychiatry, Interdisciplinary Center Psychopathology and Emotion Regulation (H.R.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Ilja Nolte
- From the Department of Epidemiology (B.S.T., T.M., N.G.A., I.N., H.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Harold Snieder
- From the Department of Epidemiology (B.S.T., T.M., N.G.A., I.N., H.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
320
|
Hou B, Jia X, Deng Z, Liu X, Liu H, Yu H, Liu S. Exploration of CYP21A2 and CYP17A1 polymorphisms and preeclampsia risk among Chinese Han population: a large-scale case-control study based on 5021 subjects. Hum Genomics 2020; 14:33. [PMID: 32977860 PMCID: PMC7517682 DOI: 10.1186/s40246-020-00286-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 06/17/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Several genome-wide association studies have identified single-nucleotide polymorphisms (SNPs), such as rs4409766, rs1004467, and rs3824755 in CYP17A1 and rs2021783 in CYP21A2, as new hypertension susceptibility genetic variants in the Chinese population. This study aimed to look into the relationship between preeclampsia (PE) and these SNPs in Chinese Han women. METHODS Overall, 5021 unrelated pregnant women were recruited, including 2002 patients with PE and 3019 normal healthy controls. The real-time PCR (TaqMan) method was applied to genotype these four polymorphisms. RESULTS A statistically obvious difference in the allelic frequencies was observed in CYP21A2 rs2021783 between cases and controls (χ2 = 7.201, Pc = 0.028 by allele), and the T allele was associated with the occurrence and development of PE (OR = 1.151, 95% CI 1.039-1.275). We also found a significant association between rs2021783 and the development of early-onset PE (Pc = 0.008 by genotype, Pc = 0.004 by allele). For rs1004467 and rs3824755, the distribution of allelic frequencies differed markedly between mild PE and control groups (χ2 = 6.843, Pc = 0.036; χ2 = 6.869, Pc = 0.036), and patients with the TT genotype of rs1004467 were less easy to develop mild PE than were those carrying the CT or CC genotype (χ2 = 7.002, Pc = 0.032, OR = 1.306, 95% CI 1.071-1.593). The GG genotype of rs3824755 appeared to a protective effect on the occurrence of mild PE (OR = 0.766, 95% CI 0.629-0.934). CONCLUSIONS CYP21A2 rs2021783 appears to be closely related to PE susceptibility, and CYP17A1 rs1004467 and rs3824755 seem to be closely associated with mild PE in Han women.
Collapse
Affiliation(s)
- Bo Hou
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuewen Jia
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Emergency Department, Shengli Oilfield Central Hospital, Dongying, China
| | - Ziwen Deng
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xin Liu
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huitang Liu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Shiguo Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China.
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
321
|
Zamanpoor M, Ghaedi H, Omrani MD. The genetic basis for the inverse relationship between rheumatoid arthritis and schizophrenia. Mol Genet Genomic Med 2020; 8:e1483. [PMID: 32965087 PMCID: PMC7667353 DOI: 10.1002/mgg3.1483] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/30/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction Rheumatoid arthritis is a common autoimmune disease and schizophrenia is a relatively common and debilitating neurological disorder. There are several common features between rheumatoid arthritis and schizophrenia. The inverse relationship between rheumatoid arthritis and schizophrenia has been replicated in several studies. Despite evidence for an inverse epidemiological relationship and negative correlations for risk between rheumatoid arthritis and schizophrenia, there are no biological data that directly support this inverse relationship. Materials and Methods’ We meta‐analyzed the genome‐wide association studies to investigate the shared association loci between rheumatoid arthritis and schizophrenia at the genome‐wide scale. Rheumatoid arthritis‐ and schizophrenia‐associated loci in most recent genome‐wide association studies of rheumatoid arthritis and schizophrenia were tested. Genetic risk score analysis was also conducted to investigate the collective contribution of schizophrenia risk loci to rheumatoid arthritis risk. Results Rheumatoid arthritis and schizophrenia meta‐genome‐wide association study showed a significant peak at the major histocompatibility complex locus on chromosome 6 in both rheumatoid arthritis‐schizophrenia meta‐genome‐wide association study and inverted meta‐genome‐wide association study datasets. Testing rheumatoid arthritis‐ and schizophrenia‐associated loci outside the human leukocyte antigen region showed no association with both rheumatoid arthritis and schizophrenia at a genome‐wide level of significance. Weighted genetic risk scores showed no evidence for a statistically significant association between rheumatoid arthritis and schizophrenia. Conclusion The finding of our study is consistent with the role of the major histocompatibility complex locus in the genetic correlation between rheumatoid arthritis and schizophrenia, and suggests that either schizophrenia has an autoimmune basis and/or rheumatoid arthritis has an active neurological component.
Collapse
Affiliation(s)
- Mansour Zamanpoor
- Medical Genetics Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Hamid Ghaedi
- Medical Genetics Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Medical Genetics Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
322
|
Vohra M, Sharma AR, Prabhu B N, Rai PS. SNPs in Sites for DNA Methylation, Transcription Factor Binding, and miRNA Targets Leading to Allele-Specific Gene Expression and Contributing to Complex Disease Risk: A Systematic Review. Public Health Genomics 2020; 23:155-170. [PMID: 32966991 DOI: 10.1159/000510253] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 07/16/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The complex genetic diversity among human populations results from an assortment of factors acting at various sequential levels, including mutations, population migrations, genetic drift, and selection. Although there are a plethora of DNA sequence variations identified through genome-wide association studies (GWAS), the challenge remains to explain the mechanisms underlying interindividual phenotypic disparity accounting for disease susceptibility. Single nucleotide polymorphisms (SNPs) present in the sites for DNA methylation, transcription factor (TF) binding, or miRNA targets can alter the gene expression. The systematic review aimed to evaluate the complex crosstalk among SNPs, miRNAs, DNA methylation, and TFs for complex multifactorial disease risk. METHODS PubMed and Scopus databases were used from inception until May 15, 2019. Initially, screening of articles involved studies assessing the interaction of SNPs with TFs, DNA methylation, or miRNAs resulting in allele-specific gene expression in complex multifactorial diseases. We also included the studies which provided experimental validation of the interaction of SNPs with each of these factors. The results from various studies on multifactorial diseases were assessed. RESULTS A total of 11 articles for SNPs interacting with DNA methylation, 30 articles for SNPs interacting with TFs, and 11 articles for SNPs in miRNA binding sites were selected. The interactions of SNPs with epigenetic factors were found to be implicated in different types of cancers, autoimmune diseases, cardiovascular diseases, diabetes, and asthma. CONCLUSION The systematic review provides evidence for the interplay between genetic and epigenetic risk factors through allele-specific gene expression in various complex multifactorial diseases.
Collapse
Affiliation(s)
- Manik Vohra
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Anu Radha Sharma
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Navya Prabhu B
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Padmalatha S Rai
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India,
| |
Collapse
|
323
|
Tang BL. SNAREs and developmental disorders. J Cell Physiol 2020; 236:2482-2504. [PMID: 32959907 DOI: 10.1002/jcp.30067] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Members of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) family mediate membrane fusion processes associated with vesicular trafficking and autophagy. SNAREs mediate core membrane fusion processes essential for all cells, but some SNAREs serve cell/tissue type-specific exocytic/endocytic functions, and are therefore critical for various aspects of embryonic development. Mutations or variants of their encoding genes could give rise to developmental disorders, such as those affecting the nervous system and immune system in humans. Mutations to components in the canonical synaptic vesicle fusion SNARE complex (VAMP2, STX1A/B, and SNAP25) and a key regulator of SNARE complex formation MUNC18-1, produce variant phenotypes of autism, intellectual disability, movement disorders, and epilepsy. STX11 and MUNC18-2 mutations underlie 2 subtypes of familial hemophagocytic lymphohistiocytosis. STX3 mutations contribute to variant microvillus inclusion disease. Chromosomal microdeletions involving STX16 play a role in pseudohypoparathyroidism type IB associated with abnormal imprinting of the GNAS complex locus. In this short review, I discuss these and other SNARE gene mutations and variants that are known to be associated with a variety developmental disorders, with a focus on their underlying cellular and molecular pathological basis deciphered through disease modeling. Possible pathogenic potentials of other SNAREs whose variants could be disease predisposing are also speculated upon.
Collapse
Affiliation(s)
- Bor L Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
324
|
Semmes EC, Shen E, Cohen JL, Zhang C, Wei Q, Hurst JH, Walsh KM. Genetic variation associated with childhood and adult stature and risk of MYCN-amplified neuroblastoma. Cancer Med 2020; 9:8216-8225. [PMID: 32945147 PMCID: PMC7643638 DOI: 10.1002/cam4.3458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/07/2020] [Accepted: 08/25/2020] [Indexed: 12/16/2022] Open
Abstract
Background Neuroblastoma is the most common pediatric solid tumor. MYCN‐amplification is an important negative prognostic indicator and inherited genetic contributions to risk are incompletely understood. Genetic determinants of stature increase risk of several adult and childhood cancers, but have not been studied in neuroblastoma despite elevated neuroblastoma incidence in children with congenital overgrowth syndromes. Methods We investigated the association between genetic determinants of height and neuroblastoma risk in 1538 neuroblastoma cases, stratified by MYCN‐amplification status, and compared to 3390 European‐ancestry controls using polygenic scores for birth length (five variants), childhood height (six variants), and adult height (413 variants). We further examined the UK Biobank to evaluate the association of known neuroblastoma risk loci and stature. Results An increase in the polygenic score for childhood stature, corresponding to a ~0.5 cm increase in pre‐pubertal height, was associated with greater risk of MYCN‐amplified neuroblastoma (OR = 1.14, P = .047). An increase in the polygenic score for adult stature, corresponding to a ~1.7 cm increase in adult height attainment, was associated with decreased risk of MYCN‐amplified neuroblastoma (OR = 0.87, P = .047). These associations persisted in case‐case analyses comparing MYCN‐amplified to MYCN‐unamplified neuroblastoma. No polygenic height scores were associated with MYCN‐unamplified neuroblastoma risk. Previously identified genome‐wide association study hits for neuroblastoma (N = 10) were significantly enriched for association with both childhood (P = 4.0 × 10−3) and adult height (P = 8.9 × 10−3) in >250 000 UK Biobank study participants. Conclusions Genetic propensity to taller childhood height and shorter adult height were associated with MYCN‐amplified neuroblastoma risk, suggesting that biological pathways affecting growth trajectories and pubertal timing may contribute to MYCN‐amplified neuroblastoma etiology.
Collapse
Affiliation(s)
- Eleanor C Semmes
- Medical Scientist Training Program, Duke University, Durham, NC, USA.,Department of Pediatrics, Children's Health and Discovery Institute, Duke University, Durham, NC, USA
| | - Erica Shen
- Division of Neuro-epidemiology, Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Jennifer L Cohen
- Division of Medical Genetics, Department of Pediatrics, Duke University, Durham, NC, USA
| | - Chenan Zhang
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Qingyi Wei
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Jillian H Hurst
- Department of Pediatrics, Children's Health and Discovery Institute, Duke University, Durham, NC, USA
| | - Kyle M Walsh
- Department of Pediatrics, Children's Health and Discovery Institute, Duke University, Durham, NC, USA.,Division of Neuro-epidemiology, Department of Neurosurgery, Duke University, Durham, NC, USA.,Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
325
|
Gray KJ, Kovacheva VP, Mirzakhani H, Bjonnes AC, Almoguera B, Wilson ML, Ingles SA, Lockwood CJ, Hakonarson H, McElrath TF, Murray JC, Norwitz ER, Karumanchi SA, Bateman BT, Keating BJ, Saxena R. Risk of pre-eclampsia in patients with a maternal genetic predisposition to common medical conditions: a case-control study. BJOG 2020; 128:55-65. [PMID: 32741103 DOI: 10.1111/1471-0528.16441] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To assess whether women with a genetic predisposition to medical conditions known to increase pre-eclampsia risk have an increased risk of pre-eclampsia in pregnancy. DESIGN Case-control study. SETTING AND POPULATION Pre-eclampsia cases (n = 498) and controls (n = 1864) in women of European ancestry from five US sites genotyped on a cardiovascular gene-centric array. METHODS Significant single-nucleotide polymorphisms (SNPs) from 21 traits in seven disease categories (cardiovascular, inflammatory/autoimmune, insulin resistance, liver, obesity, renal and thrombophilia) with published genome-wide association studies (GWAS) were used to create a genetic instrument for each trait. Multivariable logistic regression was used to test the association of each continuous scaled genetic instrument with pre-eclampsia. Odds of pre-eclampsia were compared across quartiles of the genetic instrument and evaluated for significance. MAIN OUTCOME MEASURES Genetic predisposition to medical conditions and relationship with pre-eclampsia. RESULTS An increasing burden of risk alleles for elevated diastolic blood pressure (DBP) and increased body mass index (BMI) were associated with an increased risk of pre-eclampsia (DBP, overall OR 1.11, 95% CI 1.01-1.21, P = 0.025; BMI, OR 1.10, 95% CI 1.00-1.20, P = 0.042), whereas alleles associated with elevated alkaline phosphatase (ALP) were protective (OR 0.89, 95% CI 0.82-0.97, P = 0.008), driven primarily by pleiotropic effects of variants in the FADS gene region. The effect of DBP genetic loci was even greater in early-onset pre-eclampsia cases (at <34 weeks of gestation, OR 1.30, 95% CI 1.08-1.56, P = 0.005). For other traits, there was no evidence of an association. CONCLUSIONS These results suggest that the underlying genetic architecture of pre-eclampsia may be shared with other disorders, specifically hypertension and obesity. TWEETABLE ABSTRACT A genetic predisposition to increased diastolic blood pressure and obesity increases the risk of pre-eclampsia.
Collapse
Affiliation(s)
- K J Gray
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - V P Kovacheva
- Department of Anesthesiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - H Mirzakhani
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - A C Bjonnes
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - B Almoguera
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - M L Wilson
- Department of Preventative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - S A Ingles
- Department of Preventative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - C J Lockwood
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - H Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Divisions of Human Genetics and Pulmonary Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - T F McElrath
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - J C Murray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - E R Norwitz
- Department of Obstetrics & Gynecology, Tufts Medical Center, Boston, Massachusetts, USA
| | - S A Karumanchi
- Center for Vascular Biology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - B T Bateman
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - B J Keating
- Department of Surgery and Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - R Saxena
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
326
|
Kho M, Zhao W, Ratliff SM, Ammous F, Mosley TH, Shang L, Kardia SLR, Zhou X, Smith JA. Epigenetic loci for blood pressure are associated with hypertensive target organ damage in older African Americans from the genetic epidemiology network of Arteriopathy (GENOA) study. BMC Med Genomics 2020; 13:131. [PMID: 32917208 PMCID: PMC7488710 DOI: 10.1186/s12920-020-00791-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 09/03/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Hypertension is a major modifiable risk factor for arteriosclerosis that can lead to target organ damage (TOD) of heart, kidneys, and peripheral arteries. A recent epigenome-wide association study for blood pressure (BP) identified 13 CpG sites, but it is not known whether DNA methylation at these sites is also associated with TOD. METHODS In 1218 African Americans from the Genetic Epidemiology Network of Arteriopathy (GENOA) study, a cohort of hypertensive sibships, we evaluated the associations between methylation at these 13 CpG sites measured in peripheral blood leukocytes and five TOD traits assessed approximately 5 years later. RESULTS Ten significant associations were found after adjustment for age, sex, blood cell counts, time difference between CpG and TOD measurement, and 10 genetic principal components (FDR q < 0.1): two with estimated glomerular filtration rate (eGFR, cg06690548, cg10601624), six with urinary albumin-to-creatinine ratio (UACR, cg16246545, cg14476101, cg19693031, cg06690548, cg00574958, cg22304262), and two with left ventricular mass indexed to height (LVMI, cg19693031, cg00574958). All associations with eGFR and four associations with UACR remained significant after further adjustment for body mass index (BMI), smoking status, and diabetes. We also found significant interactions between cg06690548 and BMI on UACR, and between 3 CpG sites (cg19693031, cg14476101, and cg06690548) and diabetes on UACR (FDR q < 0.1). Mediation analysis showed that 4.7% to 38.1% of the relationship between two CpG sites (cg19693031 and cg00574958) and two TOD measures (UACR and LVMI) was mediated by blood pressure (Bonferroni-corrected P < 0.05). Mendelian randomization analysis suggests that methylation at two sites (cg16246545 and cg14476101) in PHGDH may causally influence UACR. CONCLUSIONS In conclusion, we found compelling evidence for associations between arteriosclerotic traits of kidney and heart and previously identified blood pressure-associated DNA methylation sites. This study may lend insight into the role of DNA methylation in pathological mechanisms underlying target organ damage from hypertension.
Collapse
Affiliation(s)
- Minjung Kho
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Scott M. Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Farah Ammous
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Thomas H. Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS 39216 USA
| | - Lulu Shang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Xiang Zhou
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| |
Collapse
|
327
|
Amenyah SD, Ward M, McMahon A, Deane J, McNulty H, Hughes C, Strain JJ, Horigan G, Purvis J, Walsh CP, Lees-Murdock DJ. DNA methylation of hypertension-related genes and effect of riboflavin supplementation in adults stratified by genotype for the MTHFR C677T polymorphism. Int J Cardiol 2020; 322:233-239. [PMID: 32920065 DOI: 10.1016/j.ijcard.2020.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/05/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The interaction between genetic, epigenetic and environmental factors plays an important role in the aetiology of hypertension. GWAS and observational studies link the C677T polymorphism in methylenetetrahydrofolate reductase (MTHFR) with hypertension, while riboflavin, the MTHFR cofactor, has been shown to reduce blood pressure and global DNA methylation in homozygous (TT genotype) individuals. It is currently unclear whether riboflavin modulates DNA methylation of other hypertension-related genes. OBJECTIVES To compare DNA methylation of hypertension-related genes in adults stratified by MTHFR genotype and effect of riboflavin intervention in adults with the variant MTHFR 677TT genotype. METHOD Pyrosequencing was carried out for hypertension-related genes (ACE, AGTR1, GCK, GNA12, IGF2, MMP9 and NOS3) in blood samples from participants in previous trials (CC, n = 40; TT, n = 40). The effect of intervention with riboflavin (1.6 mg/d for16 weeks) or placebo on DNA methylation was investigated in adults with the variant MTHFR 677TT genotype (n = 80). RESULTS Individuals with the MTHFR 677TT v CC genotype had significantly higher average DNA methylation at NOS3 (+1.66%, P = 0.044). In response to riboflavin supplementation in TT individuals, there was an increase in average DNA methylation at IGF2 (+1.09%, P = 0.019) and a decrease at ACE (-0.44%, P = 0.021) in females only. Specific CpG sites were hypomethylated in GNA12 and hypermethylated in AGTR1. CONCLUSION This study provides the first RCT evidence that riboflavin alters DNA methylation of hypertension-related genes in adults with the MTHFR 677TT genotype, providing some insight into mechanisms linking hypertension with the genotype-specific response of BP to riboflavin.
Collapse
Affiliation(s)
- Sophia D Amenyah
- Genomic Medicine Research Group, Ulster University, Coleraine BT52 1SA, N. Ireland, UK; Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine BT52 1SA, N. Ireland, UK
| | - Mary Ward
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine BT52 1SA, N. Ireland, UK
| | - Amy McMahon
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine BT52 1SA, N. Ireland, UK
| | - Jennifer Deane
- Genomic Medicine Research Group, Ulster University, Coleraine BT52 1SA, N. Ireland, UK
| | - Helene McNulty
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine BT52 1SA, N. Ireland, UK
| | - Catherine Hughes
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine BT52 1SA, N. Ireland, UK
| | - J J Strain
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine BT52 1SA, N. Ireland, UK
| | - Geraldine Horigan
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine BT52 1SA, N. Ireland, UK
| | - John Purvis
- Department of Cardiology, Altnagelvin Area Hospital, BT47 6SB, N. Ireland, UK
| | - Colum P Walsh
- Genomic Medicine Research Group, Ulster University, Coleraine BT52 1SA, N. Ireland, UK
| | - Diane J Lees-Murdock
- Genomic Medicine Research Group, Ulster University, Coleraine BT52 1SA, N. Ireland, UK.
| |
Collapse
|
328
|
Galmés S, Serra F, Palou A. Current State of Evidence: Influence of Nutritional and Nutrigenetic Factors on Immunity in the COVID-19 Pandemic Framework. Nutrients 2020; 12:E2738. [PMID: 32911778 PMCID: PMC7551697 DOI: 10.3390/nu12092738] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/27/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
The pandemic caused by the new coronavirus has caused shock waves in many countries, producing a global health crisis worldwide. Lack of knowledge of the biological mechanisms of viruses, plus the absence of effective treatments against the disease (COVID-19) and/or vaccines have pulled factors that can compromise the proper functioning of the immune system to fight against infectious diseases into the spotlight. The optimal status of specific nutrients is considered crucial to keeping immune components within their normal activity, helping to avoid and overcome infections. Specifically, the European Food Safety Authority (EFSA) evaluated and deems six vitamins (D, A, C, Folate, B6, B12) and four minerals (zinc, iron, copper and selenium) to be essential for the normal functioning of the immune system, due to the scientific evidence collected so far. In this report, an update on the evidence of the contribution of nutritional factors as immune-enhancing aspects, factors that could reduce their bioavailability, and the role of the optimal status of these nutrients within the COVID-19 pandemic context was carried out. First, a non-systematic review of the current state of knowledge regarding the impact of an optimal nutritional status of these nutrients on the proper functioning of the immune system as well as their potential role in COVID-19 prevention/treatment was carried out by searching for available scientific evidence in PubMed and LitCovid databases. Second, a compilation from published sources and an analysis of nutritional data from 10 European countries was performed, and the relationship between country nutritional status and epidemiological COVID-19 data (available in the Worldometers database) was evaluated following an ecological study design. Furthermore, the potential effect of genetics was considered through the selection of genetic variants previously identified in Genome-Wide Association studies (GWAs) as influencing the nutritional status of these 10 considered nutrients. Therefore, access to genetic information in accessible databases (1000genomes, by Ensembl) of individuals from European populations enabled an approximation that countries might present a greater risk of suboptimal status of the nutrients studied. Results from the review approach show the importance of maintaining a correct nutritional status of these 10 nutrients analyzed for the health of the immune system, highlighting the importance of Vitamin D and iron in the context of COVID-19. Besides, the ecological study demonstrates that intake levels of relevant micronutrients-especially Vitamins D, C, B12, and iron-are inversely associated with higher COVID-19 incidence and/or mortality, particularly in populations genetically predisposed to show lower micronutrient status. In conclusion, nutrigenetic data provided by joint assessment of 10 essential nutrients for the functioning of the immune system and of the genetic factors that can limit their bioavailability can be a fundamental tool to help strengthen the immune system of individuals and prepare populations to fight against infectious diseases such as COVID-19.
Collapse
Affiliation(s)
- Sebastià Galmés
- Laboratory of Molecular Biology, Nutrition and Biotechnology, NUO Group, Universitat de les Illes Balears, 07122 Palma, Spain; (S.G.); (A.P.)
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut d’Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- Alimentómica S.L., Spin-off n.1 of the University of the Balearic Islands, 07121 Palma, Spain
| | - Francisca Serra
- Laboratory of Molecular Biology, Nutrition and Biotechnology, NUO Group, Universitat de les Illes Balears, 07122 Palma, Spain; (S.G.); (A.P.)
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut d’Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- Alimentómica S.L., Spin-off n.1 of the University of the Balearic Islands, 07121 Palma, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology, NUO Group, Universitat de les Illes Balears, 07122 Palma, Spain; (S.G.); (A.P.)
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut d’Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
| |
Collapse
|
329
|
A genome-wide association study on fish consumption in a Japanese population-the Japan Multi-Institutional Collaborative Cohort study. Eur J Clin Nutr 2020; 75:480-488. [PMID: 32895509 DOI: 10.1038/s41430-020-00702-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 07/08/2020] [Accepted: 08/04/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVE Although benefits of fish consumption for health are well known, a significant percentage of individuals dislike eating fish. Fish consumption may be influenced by genetic factors in addition to environmental factors. We conducted a genome-wide association study (GWAS) to find genetic variations that affect fish consumption in a Japanese population. METHODS We performed a two-stage GWAS on fish consumption using 13,739 discovery samples from the Japan Multi-Institutional Collaborative Cohort study, and 2845 replication samples from the other population. We used a semi-quantitative food frequency questionnaire to estimate food intake. Association of the imputed variants with fish consumption was analyzed by separate linear regression models per variant, with adjustments for age, sex, energy intake, principal component analysis components 1-10, and alcohol intake (g/day). We also performed conditional analysis. RESULTS We found 27 single nucleotide polymorphisms (SNPs) located in 12q24 and 14q32.12 that were associated with fish consumption. The 19 SNPs were located at 11 genes including six lead SNPs at the BRAP, ACAD10, ALDH2, NAA25, and HECTD4 regions on 12q24.12-13, and CCDC197 region on 14q32.12. In replication samples, all five SNPs located on chromosome 12 were replicated successfully, but the one on chromosome 14 was not. Conditional analyses revealed that the five lead variants in chromosome 12 were in fact the same signal. CONCLUSION We found that new SNPs in the 12q24 locus were related to fish intake in two Japanese populations. The associations between SNPs on chromosome 12 and fish intake were strongly confounded by drinking status.
Collapse
|
330
|
Chuluun-Erdene A, Sengeragchaa O, Altangerel TA, Sanjmyatav P, Dagdan B, Battulga S, Enkhbat L, Byambasuren N, Malchinkhuu M, Janlav M. Association of Candidate Gene Polymorphism with Metabolic Syndrome among Mongolian Subjects: A Case-Control Study. Med Sci (Basel) 2020; 8:medsci8030038. [PMID: 32887252 PMCID: PMC7563398 DOI: 10.3390/medsci8030038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022] Open
Abstract
Metabolic syndrome (MetS) is complex and determined by the interaction between genetic and environmental factors and their influence on obesity, insulin resistance, and related traits associated with diabetes and cardiovascular disease risk. Some dynamic markers, including adiponectin (ADIPOQ), brain-derived neurotrophic factor (BDNF), and lipoprotein lipase (LPL), are implicated in MetS; however, the influence of their genetic variants on MetS susceptibility varies in racial and ethnic groups. We investigated the association of single nucleotide polymorphism (SNP)-SNP interactions among nine SNPs in six genes with MetS's genetic predisposition in Mongolian subjects. A total of 160 patients with MetS for the case group and 144 healthy individuals for the control group were selected to participate in this study. Regression analysis of individual SNPs showed that the ADIPOQ + 45GG (odds ratio (OR) = 2.09, p = 0.011) and P+P+ of LPL PvuII (OR = 2.10, p = 0.038) carriers had an increased risk of MetS. Conversely, G allele of LPL S447X (OR = 0.45, p = 0.036) and PGC-1α 482Ser (OR = 0.26, p = 0.001) allele were estimated as protective factors, respectively. Moreover, a haplotype containing the G-P+-G combination was related to MetS. Significant loci were also related to body mass index (BMI), systolic blood pressure (SBP), serum high-density lipoprotein cholesterol (HDL-C), triglyceride (TG), and fasting blood glucose (FBG), adipokines, and insulin as well as insulin resistance (p < 0.05). Our results confirm that ADIPOQ + 45T > G, LPL PvII, and PGC-1α Gly482Ser loci are associated with MetS in Mongolian subjects.
Collapse
Affiliation(s)
- Ariunbold Chuluun-Erdene
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Orgil Sengeragchaa
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Tsend-Ayush Altangerel
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Purevjal Sanjmyatav
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Batnaran Dagdan
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
- Coronary Care Unit, Cardiovascular Center, The Shastin Central Hospital, Ulaanbaatar 16081, Mongolia
| | - Solongo Battulga
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Lundiamaa Enkhbat
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Nyamjav Byambasuren
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Munkhzol Malchinkhuu
- Department of Pathophysiology, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia;
| | - Munkhtstetseg Janlav
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
- Correspondence: ; Tel.: +976-9909-2287
| |
Collapse
|
331
|
Soriano-Tárraga C, Lazcano U, Giralt-Steinhauer E, Avellaneda-Gómez C, Ois Á, Rodríguez-Campello A, Cuadrado-Godia E, Gómez-González A, Fernández-Sanlés A, Elosua R, Fernández-Cadenas I, Cullell N, Montaner J, Moran S, Esteller M, Jiménez-Conde J, Roquer J. Identification of 20 novel loci associated with ischaemic stroke. Epigenome-wide association study. Epigenetics 2020; 15:988-997. [PMID: 32202197 PMCID: PMC7518691 DOI: 10.1080/15592294.2020.1746507] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/06/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023] Open
Abstract
DNA methylation is dynamic, varies throughout the life course, and its levels are influenced by lifestyle and environmental factors, as well as by genetic variation. The leading genetic variants at stroke risk loci identified to date explain roughly 1-2% of stroke heritability. Most of these single nucleotide polymorphisms are situated within a regulatory sequence marked by DNase I hypersensitivity sites, which would indicate involvement of an epigenetic mechanism. To detect epigenetic variants associated with stroke occurrence and stroke subtypes. A two-stage case-control epigenome-wide association study was designed. The discovery sample with 401 samples included 218 ischaemic stroke (IS) patients, assessed at Hospital del Mar (Barcelona, Spain) and 183 controls from the REGICOR cohort. In two independent samples (N = 226 and N = 166), we replicated 22 CpG sites differentially methylated in IS in 21 loci, including 2 CpGs in locus ZFHX3, which includes known genetic variants associated with stroke. The pathways associated with these loci are inflammation and angiogenesis. The meta-analysis identified 384 differentially methylated CpGs, including loci of known stroke and vascular risk genetic variants, enriched by loci involved in lipid metabolism, adipogenesis, circadian clock, and glycolysis pathways. We identified a set of 22 CpGs in 21 loci associated with IS. Our analysis suggests that DNA methylation changes may contribute to orchestrating gene expression that contributes to IS.
Collapse
Affiliation(s)
- Carolina Soriano-Tárraga
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Uxue Lazcano
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Eva Giralt-Steinhauer
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Carla Avellaneda-Gómez
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Ángel Ois
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Ana Rodríguez-Campello
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Elisa Cuadrado-Godia
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Alejandra Gómez-González
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Alba Fernández-Sanlés
- Cardiovascular Epidemiology and Genetics Research Group, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics Research Group, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, Barcelona, Spain
- Medicine Department, Medical School, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Israel Fernández-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Natalia Cullell
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Neurology, Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d’Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Terrassa, Spain
- Institute de Biomedicine of Seville, IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville & Department of Neurology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Sebastian Moran
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet, Barcelona, Spain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
| | - Jordi Jiménez-Conde
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Jaume Roquer
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d’Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
332
|
Miller JB, Ward E, Staley LA, Stevens J, Teerlink CC, Tavana JP, Cloward M, Page M, Dayton L, Cannon-Albright LA, Kauwe JSK. Identification and genomic analysis of pedigrees with exceptional longevity identifies candidate rare variants. Neurobiol Dis 2020; 143:104972. [PMID: 32574725 PMCID: PMC7461696 DOI: 10.1016/j.nbd.2020.104972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/05/2020] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Longevity as a phenotype entails living longer than average and typically includes living without chronic age-related diseases. Recently, several common genetic components to longevity have been identified. This study aims to identify additional genetic variants associated with longevity using unique and powerful analyses of pedigrees with a statistical excess of healthy elderly individuals identified in the Utah Population Database (UPDB). METHODS From an existing biorepository of Utah pedigrees, six independent cousin pairs were selected from four extended pedigrees that exhibited an excess of healthy elderly individuals; whole exome sequencing (WES) was performed on two elderly individuals from each pedigree who were either first cousins or first cousins once removed. Rare (<.01 population frequency) variants shared by at least one elderly cousin pair in a region likely to be identical by descent were identified as candidates. Ingenuity Variant Analysis was used to prioritize putative causal variants based on quality control, frequency, and gain or loss of function. The variant frequency was compared in healthy cohorts and in an Alzheimer's disease cohort. Remaining variants were filtered based on their presence in genes reported to have an effect on the aging process, aging of cells, or the longevity process. Validation of these candidate variants included tests of segregation on other elderly relatives. RESULTS Fifteen rare candidate genetic variants spanning 17 genes shared within cousins were identified as having passed prioritization criteria. Of those variants, six were present in genes that are known or predicted to affect the aging process: rs78408340 (PAM), rs112892337 (ZFAT), rs61737629 (ESPL1), rs141903485 (CEBPE), rs144369314 (UTP4), and rs61753103 (NUP88 and RABEP1). ESPL1 rs61737629 and CEBPE rs141903485 show additional evidence of segregation with longevity in expanded pedigree analyses (p-values = .001 and .0001, respectively). DISCUSSION This unique pedigree analysis efficiently identified several novel rare candidate variants that may affect the aging process and added support to seven genes that likely contribute to longevity. Further analyses showed evidence for segregation for two rare variants, ESPL1 rs61737629 and CEBPE rs141903485, in the original longevity pedigrees in which they were initially observed. These candidate genes and variants warrant further investigation.
Collapse
Affiliation(s)
- Justin B Miller
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Elizabeth Ward
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Lyndsay A Staley
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Jeffrey Stevens
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Craig C Teerlink
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Justina P Tavana
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Matthew Cloward
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Madeline Page
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Louisa Dayton
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Lisa A Cannon-Albright
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
333
|
Poveda A, Atabaki‐Pasdar N, Ahmad S, Hallmans G, Renström F, Franks PW. Association of Established Blood Pressure Loci With 10-Year Change in Blood Pressure and Their Ability to Predict Incident Hypertension. J Am Heart Assoc 2020; 9:e014513. [PMID: 32805198 PMCID: PMC7660819 DOI: 10.1161/jaha.119.014513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 06/11/2020] [Indexed: 01/11/2023]
Abstract
Background Genome-wide association studies have identified >1000 genetic variants cross-sectionally associated with blood pressure variation and prevalent hypertension. These discoveries might aid the early identification of subpopulations at risk of developing hypertension or provide targets for drug development, amongst other applications. The aim of the present study was to analyze the association of blood pressure-associated variants with long-term changes (10 years) in blood pressure and also to assess their ability to predict hypertension incidence compared with traditional risk variables in a Swedish population. Methods and Results We constructed 6 genetic risk scores (GRSs) by summing the dosage of the effect allele at each locus of genetic variants previously associated with blood pressure traits (systolic blood pressure GRS (GRSSBP): 554 variants; diastolic blood pressure GRS (GRSDBP): 481 variants; mean arterial pressure GRS (GRSMAP): 20 variants; pulse pressure GRS (GRSPP): 478 variants; hypertension GRS (GRSHTN): 22 variants; combined GRS (GRScomb): 1152 variants). Each GRS was longitudinally associated with its corresponding blood pressure trait, with estimated effects per GRS SD unit of 0.50 to 1.21 mm Hg for quantitative traits and odds ratios (ORs) of 1.10 to 1.35 for hypertension incidence traits. The GRScomb was also significantly associated with hypertension incidence defined according to European guidelines (OR, 1.22 per SD; 95% CI, 1.10‒1.35) but not US guidelines (OR, 1.11 per SD; 95% CI, 0.99‒1.25) while controlling for traditional risk factors. The addition of GRScomb to a model containing traditional risk factors only marginally improved discrimination (Δarea under the ROC curve = 0.001-0.002). Conclusions GRSs based on discovered blood pressure-associated variants are associated with long-term changes in blood pressure traits and hypertension incidence, but the inclusion of genetic factors in a model composed of conventional hypertension risk factors did not yield a material increase in predictive ability.
Collapse
Affiliation(s)
- Alaitz Poveda
- Genetic and Molecular Epidemiology UnitDepartment of Clinical SciencesLund University Diabetes CentreLund UniversityMalmöSweden
| | - Naeimeh Atabaki‐Pasdar
- Genetic and Molecular Epidemiology UnitDepartment of Clinical SciencesLund University Diabetes CentreLund UniversityMalmöSweden
| | - Shafqat Ahmad
- Preventive Medicine DivisionBrigham and Women's HospitalHarvard Medical SchoolBostonMA
- Department of Medical SciencesMolecular EpidemiologyUppsala UniversityUppsalaSweden
| | - Göran Hallmans
- Section for Nutritional ResearchDepartment of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
| | - Frida Renström
- Genetic and Molecular Epidemiology UnitDepartment of Clinical SciencesLund University Diabetes CentreLund UniversityMalmöSweden
- Section for Nutritional ResearchDepartment of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
- Division of Endocrinology and DiabetesCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Paul W. Franks
- Genetic and Molecular Epidemiology UnitDepartment of Clinical SciencesLund University Diabetes CentreLund UniversityMalmöSweden
- Section for Nutritional ResearchDepartment of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
- Department of NutritionHarvard Chan School of Public HealthBostonMA
| |
Collapse
|
334
|
A higher level of serum furin indicates a higher risk of microalbuminuria: results from a longitudinal study in Chinese adults. Clin Exp Nephrol 2020; 24:885-892. [PMID: 32770419 DOI: 10.1007/s10157-020-01912-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 06/01/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Furin, a key enzyme of natriuretic peptide system, has been suggested to play a role in microalbuminuria, but the association between furin and microalbuminuria has been scarcely studied. METHODS Leveraging a longitudinal cohort of Chinese adults who had urinary albumin measured twice 4 years apart, we examined the cross-sectional and prospective associations of baseline serum furin with microalbuminuria, adjusting for age, sex, education level, smoking, drinking, obesity, blood pressure, glucose, lipids, and antihypertensive medications. RESULTS The cross-sectional analysis in 2175 participants (53 ± 10 years, 38% men) found that a 10-time higher level of serum furin was significantly associated with a 64% higher risk of having microalbuminuria (OR = 1.64, P = 0.005). The longitudinal analysis found a positive association between baseline serum furin and dynamic elevation of albumin excretion during follow-up. The prospective analysis in 1357 participants free of microalbuminuria at baseline found that a 10-time higher level of serum furin at baseline was significantly associated with a 1.28-time higher risk of developing microalbuminuria 4 years later (OR = 2.28, P < 0.001). CONCLUSIONS A higher level of serum furin at baseline predicted an increased risk of developing microalbuminuria in Chinese adults. These findings indicate that furin might be a predictor or a risk factor for microalbuminuria but the causality still needs more investigations.
Collapse
|
335
|
Kessler T, Schunkert H, von Hundelshausen P. Novel Approaches to Fine-Tune Therapeutic Targeting of Platelets in Atherosclerosis: A Critical Appraisal. Thromb Haemost 2020; 120:1492-1504. [PMID: 32772352 DOI: 10.1055/s-0040-1714352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The pathogenesis of atherosclerotic vascular disease is driven by a multitude of risk factors intertwining metabolic and inflammatory pathways. Increasing knowledge about platelet biology sheds light on how platelets take part in these processes from early to later stages of plaque development. Recent insights from experimental studies and mouse models substantiate platelets as initiators and amplifiers in atherogenic leukocyte recruitment. These studies are complemented by results from genetics studies shedding light on novel molecular mechanisms which provide an interesting prospect as novel targets. For instance, experimental studies provide further details how platelet-decorated von Willebrand factor tethered to activated endothelial cells plays a role in atherogenic monocyte recruitment. Novel aspects of platelets as atherogenic inductors of neutrophil extracellular traps and particularities in signaling pathways such as cyclic guanosine monophosphate and the inhibitory adaptor molecule SHB23/LNK associating platelets with atherogenesis are shared. In summary, it was our intention to balance insights from recent experimental data that support a plausible role for platelets in atherogenesis against a paucity of clinical evidence needed to validate this concept in humans.
Collapse
Affiliation(s)
- Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany
| | - Philipp von Hundelshausen
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany.,Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, Klinikum der Universität, Ludwig-Maximilians-Universität, Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
336
|
Abstract
Previous genetic association studies have failed to identify loci robustly associated with sepsis, and there have been no published genetic association studies or polygenic risk score analyses of patients with septic shock, despite evidence suggesting genetic factors may be involved. We systematically collected genotype and clinical outcome data in the context of a randomized controlled trial from patients with septic shock to enrich the presence of disease-associated genetic variants. We performed genomewide association studies of susceptibility and mortality in septic shock using 493 patients with septic shock and 2442 population controls, and polygenic risk score analysis to assess genetic overlap between septic shock risk/mortality with clinically relevant traits. One variant, rs9489328, located in AL589740.1 noncoding RNA, was significantly associated with septic shock (p = 1.05 × 10-10); however, it is likely a false-positive. We were unable to replicate variants previously reported to be associated (p < 1.00 × 10-6 in previous scans) with susceptibility to and mortality from sepsis. Polygenic risk scores for hematocrit and granulocyte count were negatively associated with 28-day mortality (p = 3.04 × 10-3; p = 2.29 × 10-3), and scores for C-reactive protein levels were positively associated with susceptibility to septic shock (p = 1.44 × 10-3). Results suggest that common variants of large effect do not influence septic shock susceptibility, mortality and resolution; however, genetic predispositions to clinically relevant traits are significantly associated with increased susceptibility and mortality in septic individuals.
Collapse
|
337
|
Mealer RG, Jenkins BG, Chen CY, Daly MJ, Ge T, Lehoux S, Marquardt T, Palmer CD, Park JH, Parsons PJ, Sackstein R, Williams SE, Cummings RD, Scolnick EM, Smoller JW. The schizophrenia risk locus in SLC39A8 alters brain metal transport and plasma glycosylation. Sci Rep 2020; 10:13162. [PMID: 32753748 PMCID: PMC7403432 DOI: 10.1038/s41598-020-70108-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022] Open
Abstract
A common missense variant in SLC39A8 is convincingly associated with schizophrenia and several additional phenotypes. Homozygous loss-of-function mutations in SLC39A8 result in undetectable serum manganese (Mn) and a Congenital Disorder of Glycosylation (CDG) due to the exquisite sensitivity of glycosyltransferases to Mn concentration. Here, we identified several Mn-related changes in human carriers of the common SLC39A8 missense allele. Analysis of structural brain MRI scans showed a dose-dependent change in the ratio of T2w to T1w signal in several regions. Comprehensive trace element analysis confirmed a specific reduction of only serum Mn, and plasma protein N-glycome profiling revealed reduced complexity and branching. N-glycome profiling from two individuals with SLC39A8-CDG showed similar but more severe alterations in branching that improved with Mn supplementation, suggesting that the common variant exists on a spectrum of hypofunction with potential for reversibility. Characterizing the functional impact of this variant will enhance our understanding of schizophrenia pathogenesis and identify novel therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Robert G Mealer
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA.
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Bruce G Jenkins
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Chia-Yen Chen
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark J Daly
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Sylvain Lehoux
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Thorsten Marquardt
- Klinik und Poliklinik für Kinder- und Jugendmedizin-Allgemeine Pädiatrie, Universitätsklinikum Münster, Münster, Germany
| | - Christopher D Palmer
- Laboratory of Inorganic and Nuclear Chemistry, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, Albany, NY, USA
| | - Julien H Park
- Klinik und Poliklinik für Kinder- und Jugendmedizin-Allgemeine Pädiatrie, Universitätsklinikum Münster, Münster, Germany
| | - Patrick J Parsons
- Laboratory of Inorganic and Nuclear Chemistry, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, Albany, NY, USA
| | - Robert Sackstein
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Sarah E Williams
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Richard D Cummings
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Edward M Scolnick
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
| |
Collapse
|
338
|
Kunnas T, Nikkari ST. Positive Association of the JAG1 rs1327235 Genotype with Coronary Artery Disease in Men, the Tampere Adult Population Cardiovascular Risk Study. Genet Test Mol Biomarkers 2020; 24:681-684. [PMID: 32721243 DOI: 10.1089/gtmb.2020.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Aims: The intronic single nucleotide polymorphism rs1327235 (A>G) close to the JAG gene has been implicated to be involved in blood pressure physiology in a genome-wide association study. We wanted to study whether it was associated with hypertension and coronary artery disease (CAD) in the Tampere adult population cardiovascular risk study. Materials and Methods: We analyzed a Finnish periodic health examination cohort of 191 men with diagnosed hypertension and 295 controls. Samples were genotyped for the JAG1 rs1327235 polymorphism using Competitive Allelic Specific PCR (KASP). The incidence of CAD was determined by self-report and the National Hospital Discharge Registry (HILMO). Results: There was no association between the JAG1 rs1327235 genotypes with hypertension at the age of 50 years. However, when the subjects were followed to the age of 60 years, those with the genotype GG had a higher prevalence of CAD (17.9%), compared with the A-allele (9.7%) (p = 0.036). When prevalence of CAD was adjusted by body mass index and total cholesterol, the OR for GG genotype was 2.19 (p = 0.029, confidence interval 1.084 - 4.429) compared with A-allele carriers. In addition, the GG genotype was associated with higher total cholesterol and low-density lipoprotein-cholesterol values, compared with the A-allele. Conclusions: Our findings suggest that the variations in JAG1 rs1327235 may be involved in CAD and cholesterol metabolism.
Collapse
Affiliation(s)
- Tarja Kunnas
- Department of Medical Biochemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Laboratories, Tampere, Finland
| | - Seppo T Nikkari
- Department of Medical Biochemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Laboratories, Tampere, Finland
| |
Collapse
|
339
|
Nandakumar P, Lee D, Hoffmann TJ, Ehret GB, Arking D, Ranatunga D, Li M, Grove ML, Boerwinkle E, Schaefer C, Kwok PY, Iribarren C, Risch N, Chakravarti A. Analysis of putative cis-regulatory elements regulating blood pressure variation. Hum Mol Genet 2020; 29:1922-1932. [PMID: 32436959 PMCID: PMC7372556 DOI: 10.1093/hmg/ddaa098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/29/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022] Open
Abstract
Hundreds of loci have been associated with blood pressure (BP) traits from many genome-wide association studies. We identified an enrichment of these loci in aorta and tibial artery expression quantitative trait loci in our previous work in ~100 000 Genetic Epidemiology Research on Aging study participants. In the present study, we sought to fine-map known loci and identify novel genes by determining putative regulatory regions for these and other tissues relevant to BP. We constructed maps of putative cis-regulatory elements (CREs) using publicly available open chromatin data for the heart, aorta and tibial arteries, and multiple kidney cell types. Variants within these regions may be evaluated quantitatively for their tissue- or cell-type-specific regulatory impact using deltaSVM functional scores, as described in our previous work. We aggregate variants within these putative CREs within 50 Kb of the start or end of 'expressed' genes in these tissues or cell types using public expression data and use deltaSVM scores as weights in the group-wise sequence kernel association test to identify candidates. We test for association with both BP traits and expression within these tissues or cell types of interest and identify the candidates MTHFR, C10orf32, CSK, NOV, ULK4, SDCCAG8, SCAMP5, RPP25, HDGFRP3, VPS37B and PPCDC. Additionally, we examined two known QT interval genes, SCN5A and NOS1AP, in the Atherosclerosis Risk in Communities Study, as a positive control, and observed the expected heart-specific effect. Thus, our method identifies variants and genes for further functional testing using tissue- or cell-type-specific putative regulatory information.
Collapse
Affiliation(s)
- Priyanka Nandakumar
- Department of Genetic Medicine, McKusick-Nathans Institute, Baltimore, MD 21205, USA
| | - Dongwon Lee
- Department of Genetic Medicine, McKusick-Nathans Institute, Baltimore, MD 21205, USA
- Center for Human Genetics and Genomics, NYU School of Medicine, New York, NY 10016, USA
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Thomas J Hoffmann
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Georg B Ehret
- Department of Genetic Medicine, McKusick-Nathans Institute, Baltimore, MD 21205, USA
- Center for Human Genetics and Genomics, NYU School of Medicine, New York, NY 10016, USA
- Cardiology, Department of Specialties of Internal Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Dan Arking
- Department of Genetic Medicine, McKusick-Nathans Institute, Baltimore, MD 21205, USA
| | - Dilrini Ranatunga
- Kaiser Permanente Northern California Division of Research, Oakland, California 94612 USA
| | - Man Li
- Division of Nephrology, Department of Human Genetics, University of Utah, Salt Lake City, Utah 84132, USA
| | - Megan L Grove
- Human Genetics Center, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Catherine Schaefer
- Kaiser Permanente Northern California Division of Research, Oakland, California 94612 USA
| | - Pui-Yan Kwok
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Carlos Iribarren
- Kaiser Permanente Northern California Division of Research, Oakland, California 94612 USA
| | - Neil Risch
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
- Kaiser Permanente Northern California Division of Research, Oakland, California 94612 USA
| | - Aravinda Chakravarti
- Department of Genetic Medicine, McKusick-Nathans Institute, Baltimore, MD 21205, USA
- Center for Human Genetics and Genomics, NYU School of Medicine, New York, NY 10016, USA
| |
Collapse
|
340
|
Human essential hypertension: no significant association of polygenic risk scores with antihypertensive drug responses. Sci Rep 2020; 10:11940. [PMID: 32686723 PMCID: PMC7371738 DOI: 10.1038/s41598-020-68878-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/30/2020] [Indexed: 11/08/2022] Open
Abstract
Polygenic risk scores (PRSs) for essential hypertension, calculated from > 900 genomic loci, were recently found to explain a significant fraction of hypertension heritability and complications. To investigate whether variation of hypertension PRS also captures variation of antihypertensive drug responsiveness, we calculated two different PRSs for both systolic and diastolic blood pressure: one based on the top 793 independent hypertension-associated single nucleotide polymorphisms and another based on over 1 million genome-wide variants. Using our pharmacogenomic GENRES study comprising four different antihypertensive monotherapies (n ~ 200 for all drugs), we identified a weak, but (after Bonferroni correction) statistically nonsignificant association of higher genome-wide PRSs with weaker response to a diuretic. In addition, we noticed a correlation between high genome-wide PRS and electrocardiographic left ventricular hypertrophy. Finally, using data of the Finnish arm of the LIFE study (n = 346), we found that PRSs for systolic blood pressure were slightly higher in patients with drug-resistant hypertension than in those with drug-controlled hypertension (p = 0.03, not significant after Bonferroni correction). In conclusion, our results indicate that patients with elevated hypertension PRSs may be predisposed to difficult-to-control hypertension and complications thereof. No general association between a high PRS and less efficient drug responsiveness was noticed.
Collapse
|
341
|
Zhloba AA, Subbotina TF. [The evaluation of homoarginine and folic acid in patients with arterial hypertension.]. Klin Lab Diagn 2020; 65:474-481. [PMID: 32762188 DOI: 10.18821/0869-2084-2020-65-8-474-481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
According to current data, a low level of folic acid (FA) contributes to the progression of arterial hypertension (AH), affecting the metabolism of cells that are involved in the vascular tone regulation, such as hypothalamic astrocytes of nervous tissue. It is also known that the level of FA in the nervous tissue and cerebrospinal fluid is 2-3 times higher than in plasma. There is another metabolic marker of cardiovascular diseases, the level of plasma homoarginine (hArg). The decrease in the level of plasma hArg is also known as a diagnostic sign. In our study, we established that in patients with AH (n = 60), the level of hArg was almost 2 times lower than in healthy individuals, and in 75% of cases the rate was below 1.80 μM. The insufficiency of FA taking into account its low level in plasma FA, as well as the level of total homocysteine (tHcy) higher than 10.9 μM, was observed in 78% of patients. HArg levels at values less than 1.80 μM corresponded to a statistically significant decrease in FA when its content was less than 13.5 nM. This relationship (r = 0.63, p = 0.020) appears in patients with AH, regardless of the number and severity of target organ damage (TOD). FA and hArg as metabolic markers exhibit various diagnostic capabilities when comparing subgroups of patients without TOD and with multiple TOD. Significant differences fared at an acceptable level (p = 0.007) only for the hArg levels, while for the FA concentrations there was only a trend to decrease. It is possible that metabolic disturbances in the central nervous system that are associated with the necessary to maintain high FA concentration contribute to the development of hypertensive status. The causal relationship of a parallel decrease in hArg and FA levels in patients with AH requires further research.
Collapse
Affiliation(s)
- A A Zhloba
- Pavlov First Saint Petersburg State Medical University of Minzdrav of Russia, 197022, Saint-Petersburg, Russia
| | - T F Subbotina
- Pavlov First Saint Petersburg State Medical University of Minzdrav of Russia, 197022, Saint-Petersburg, Russia
| |
Collapse
|
342
|
Association of SBP and BMI with cognitive and structural brain phenotypes in UK Biobank. J Hypertens 2020; 38:2482-2489. [DOI: 10.1097/hjh.0000000000002579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
343
|
Gorabi AM, Penson PE, Banach M, Motallebnezhad M, Jamialahmadi T, Sahebkar A. Epigenetic control of atherosclerosis via DNA methylation: A new therapeutic target? Life Sci 2020; 253:117682. [PMID: 32387418 DOI: 10.1016/j.lfs.2020.117682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a disease in which lipid-laden plaques are developed inside the vessel walls of arteries. The immune system is activated, resulting in inflammation and oxidative stress. Endothelial cells (ECs) are activated, arterial smooth muscle cells (SMCs) proliferate, macrophages are activated, and foam cells are developed, leading to dysfunctional ECs. Epigenetic regulatory mechanisms, including DNA methylation, histone modifications, and microRNAs are involved in the modulation of genes that play distinct roles in several aspects of cell biology and physiology, hence linking environmental stimuli to gene regulation. Recent research has investigated the involvement of DNA methylation in the etiopathogenesis of atherosclerosis, and several studies have documented the role of this mechanism in various aspects of the disease. Regulation of DNA methylation plays a critical role in the integrity of ECs, SMC proliferation and formation of atherosclerotic lesions. In this review, we seek to clarify the role of DNA methylation in the development of atherosclerosis through different mechanisms.
Collapse
Affiliation(s)
- Armita Mahdavi Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Peter E Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Morteza Motallebnezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
344
|
Impaired iloprost-induced platelet inhibition and phosphoproteome changes in patients with confirmed pseudohypoparathyroidism type Ia, linked to genetic mutations in GNAS. Sci Rep 2020; 10:11389. [PMID: 32647264 PMCID: PMC7347634 DOI: 10.1038/s41598-020-68379-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/22/2020] [Indexed: 11/16/2022] Open
Abstract
Patients diagnosed with pseudohypoparathyroidism type Ia (PHP Ia) suffer from hormonal resistance and abnormal postural features, in a condition classified as Albright hereditary osteodystrophy (AHO) syndrome. This syndrome is linked to a maternally inherited mutation in the GNAS complex locus, encoding for the GTPase subunit Gsα. Here, we investigated how platelet phenotype and omics analysis can assist in the often difficult diagnosis. By coupling to the IP receptor, Gsα induces platelet inhibition via adenylyl cyclase and cAMP-dependent protein kinase A (PKA). In platelets from seven patients with suspected AHO, one of the largest cohorts examined, we studied the PKA-induced phenotypic changes. Five patients with a confirmed GNAS mutation, displayed impairments in Gsα-dependent VASP phosphorylation, aggregation, and microfluidic thrombus formation. Analysis of the platelet phosphoproteome revealed 2,516 phosphorylation sites, of which 453 were regulated by Gsα-PKA. Common changes in the patients were: (1) a joint panel of upregulated and downregulated phosphopeptides; (2) overall PKA dependency of the upregulated phosphopeptides; (3) links to key platelet function pathways. In one patient with GNAS mutation, diagnosed as non-AHO, the changes in platelet phosphoproteome were reversed. This combined approach thus revealed multiple phenotypic and molecular biomarkers to assist in the diagnosis of suspected PHP Ia.
Collapse
|
345
|
A new approach to identifying hypertension-associated genes in the mesenteric artery of spontaneously hypertensive rats and stroke-prone spontaneously hypertensive rats. J Hypertens 2020; 37:1644-1656. [PMID: 30882592 PMCID: PMC6615961 DOI: 10.1097/hjh.0000000000002083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Supplemental Digital Content is available in the text Objective: Hypertension is one of the most prevalent diseases in humans who live a modern lifestyle. Alongside more effective care, clarification of the genetic background of hypertension is urgently required. Gene expression in mesenteric resistance arteries of spontaneously hypertensive rats (SHR), stroke-prone SHR (SHRSP) and two types of renal hypertensive Wistar Kyoto rats (WKY), two kidneys and one clip renal hypertensive rat (2K1C) and one kidney and one clip renal hypertensive rat (1K1C), was compared using DNA microarrays. Methods: We used a simultaneous equation and comparative selection method to identify genes associated with hypertension using the Reactome analysis tool and GenBank database. Results: The expression of 298 genes was altered between SHR and WKY (44 upregulated and 254 downregulated), while the expression of 290 genes was altered between SHRSP and WKY (83 upregulated and 207 downregulated). For SHRSP versus SHR, the expression of 60 genes was altered (36 upregulated and 24 downregulated). Several genes expressed in SHR and SHRSP were also expressed in the renovascular hypertensive 2K1C and 1K1C rats, indicative of the existence of hyper-renin and/or hypervolemic pathophysiological changes in SHR and SHRSP. Conclusion: The overexpression of Kcnq1, Crlf1, Alb and Xirp1 and the inhibition of Galr2, Kcnh1, Ache, Chrm2 and Slc5a7 expression may indicate that a relationship exists between these genes and the cause and/or worsening of hypertension in SHR and SHRSP.
Collapse
|
346
|
Lin J, Susztak K. Complexities of Understanding Function from CKD-Associated DNA Variants. Clin J Am Soc Nephrol 2020; 15:1028-1040. [PMID: 32513823 PMCID: PMC7341770 DOI: 10.2215/cjn.15771219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Genome-wide association studies (GWASs) have facilitated the unbiased discovery of hundreds of genomic loci associated with CKD and kidney function. The vast majority of disease-associated DNA variants are noncoding. Those that are causal in CKD pathogenesis likely modulate transcription of target genes in a cell type-specific manner. To gain novel biological insights into mechanisms driving the development of CKD, the causal variants (which are usually not the most significant variant reported in a GWAS), their target genes, and causal cell types need to be identified. This functional validation requires a large number of new data sets, complex bioinformatics analyses, and experimental cellular and in vivo studies. Here, we review the basic principles and some of the current approaches being leveraged to assign functional significance to a genotype-phenotype association.
Collapse
Affiliation(s)
- Jennie Lin
- Division of Nephrology and Hypertension, Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Northwestern University, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Genetics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
347
|
Morrison J, Knoblauch N, Marcus JH, Stephens M, He X. Mendelian randomization accounting for correlated and uncorrelated pleiotropic effects using genome-wide summary statistics. Nat Genet 2020; 52:740-747. [PMID: 32451458 PMCID: PMC7343608 DOI: 10.1038/s41588-020-0631-4] [Citation(s) in RCA: 349] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 04/20/2020] [Indexed: 11/09/2022]
Abstract
Mendelian randomization (MR) is a valuable tool for detecting causal effects by using genetic variant associations. Opportunities to apply MR are growing rapidly with the increasing number of genome-wide association studies (GWAS). However, existing MR methods rely on strong assumptions that are often violated, leading to false positives. Correlated horizontal pleiotropy, which arises when variants affect both traits through a heritable shared factor, remains a particularly challenging problem. We propose a new MR method, Causal Analysis Using Summary Effect estimates (CAUSE), that accounts for correlated and uncorrelated horizontal pleiotropic effects. We demonstrate, in simulations, that CAUSE avoids more false positives induced by correlated horizontal pleiotropy than other methods. Applied to traits studied in recent GWAS studies, we find that CAUSE detects causal relationships that have strong literature support and avoids identifying most unlikely relationships. Our results suggest that shared heritable factors are common and may lead to many false positives using alternative methods.
Collapse
Affiliation(s)
- Jean Morrison
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | | | - Joseph H Marcus
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Matthew Stephens
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Department of Statistics, University of Chicago, Chicago, IL, USA
| | - Xin He
- Department of Human Genetics, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
348
|
Smart analysis of a ternary mixture of amlodipine, hydrochlorothiazide and telmisartan by manipulation of UV spectra: Development, validation and application to formulations. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
349
|
Yan S, Lian XQ, Wang SB, Wang H, Wang LS. Hyperplasia suppressor gene polymorphisms and essential hypertension: a case-control association study in a central Han Chinese population. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:1886-1896. [PMID: 32782719 PMCID: PMC7414465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/11/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND HSG (hyperplasia suppressor gene, also named Mitofusion-2, Mfn-2) gene polymorphisms have been studied as a candidate gene in essential hypertension, but no clear consensus has been reached in the Chinese population. To systematically explore their possible association, a case-control study was conducted in a central Chinese population. METHODS AND RESULTS We recruited 402 EH patients and 267 normotensive (NT) control subjects. A total of 6 tag SNPs of HSG gene were genotyped successfully by TaqMan assay. The results showed that genotype distribution and the allelic frequency of rs873457, rs2236384, rs4846085, and rs1474868 in the EH and NT groups were significantly different (P < 0.05), although those of rs2295281 and rs17037564 were not. rs2336384, rs873457, rs4846085 and rs1474868 were also closely associated with EH under the dominant genetic model (P < 0.05). Gender-based subgroup analyses showed that significant associations between rs873457, rs2336384, rs4846085, and rs1474868 and EH could be found in males, but not in females. Haplotype analysis indicated that the C-G-T-T-T-G haplotype was positively correlated with EH. CONCLUSION Our study suggested that HSG gene polymorphisms were significantly associated with EH in a central Han Chinese population, especially in male subjects.
Collapse
Affiliation(s)
- Shan Yan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu Province, China
- Department of Cardiology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical UniversityHuaian 223300, Jiangsu Province, China
| | - Xiao-Qing Lian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu Province, China
| | - Si-Bo Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu Province, China
| | - Hao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu Province, China
| | - Lian-Sheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu Province, China
| |
Collapse
|
350
|
Bardsley EN, Paterson DJ. Neurocardiac regulation: from cardiac mechanisms to novel therapeutic approaches. J Physiol 2020; 598:2957-2976. [PMID: 30307615 PMCID: PMC7496613 DOI: 10.1113/jp276962] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/02/2018] [Indexed: 12/15/2022] Open
Abstract
Cardiac sympathetic overactivity is a well-established contributor to the progression of neurogenic hypertension and heart failure, yet the underlying pathophysiology remains unclear. Recent studies have highlighted the importance of acutely regulated cyclic nucleotides and their effectors in the control of intracellular calcium and exocytosis. Emerging evidence now suggests that a significant component of sympathetic overactivity and enhanced transmission may arise from impaired cyclic nucleotide signalling, resulting from compromised phosphodiesterase activity, as well as alterations in receptor-coupled G-protein activation. In this review, we address some of the key cellular and molecular pathways that contribute to sympathetic overactivity in hypertension and discuss their potential for therapeutic targeting.
Collapse
Affiliation(s)
- E. N. Bardsley
- Wellcome Trust OXION Initiative in Ion Channels and DiseaseOxfordUK
- Burdon Sanderson Cardiac Science Centre, Department of PhysiologyAnatomy and Genetics, University of OxfordOxfordOX1 3PTUK
| | - D. J. Paterson
- Wellcome Trust OXION Initiative in Ion Channels and DiseaseOxfordUK
- Burdon Sanderson Cardiac Science Centre, Department of PhysiologyAnatomy and Genetics, University of OxfordOxfordOX1 3PTUK
| |
Collapse
|