301
|
Estes C, Chan HLY, Chien RN, Chuang W, Fung J, Goh GBB, Hu TH, Huang J, Jang BK, Jun DW, Kao JH, Lee J, Lin H, Razavi‐Shearer K, Seto W, Wong GL‐H, Wong VW‐S, Razavi H. Modelling NAFLD disease burden in four Asian regions-2019-2030. Aliment Pharmacol Ther 2020; 51:801-811. [PMID: 32133676 PMCID: PMC7154715 DOI: 10.1111/apt.15673] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/27/2020] [Accepted: 02/06/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) account for an increasing proportion of liver disease in the Asia-Pacific region. Many areas in the region are experiencing epidemics of metabolic syndrome among rapidly ageing populations. AIMS To estimate using modelling the growth in NAFLD populations, including cases with significant fibrosis that are most likely to experience advanced liver disease and related mortality. METHODS A disease progression model was used to summarise and project fibrosis progression among the NAFLD populations of Hong Kong, Singapore, South Korea and Taiwan. For each area, changes in the adult prevalence of obesity was used to extrapolate long-term trends in NAFLD incidence. RESULTS In the areas studied, prevalent NAFLD cases were projected to increase 6%-20% during 2019-2030, while prevalent NASH cases increase 20%-35%. Incident cases of hepatocellular carcinoma are projected to increase by 65%-85%, while incident decompensated cirrhosis cases increase 65%-100% by 2030. Likewise, NAFLD-related mortality is projected to increase between 65% and 100% from 2019 to 2030. NAFLD disease burden is expected to increase alongside rising trends in metabolic syndrome and obesity among populations in the region. This leads to more cases of advanced liver disease and associated mortality. CONCLUSIONS Preventing the growth of diabetic and obese populations will be a key factor in reducing ongoing increases in NAFLD-related disease burden in the Asia-Pacific region.
Collapse
|
302
|
Tietge UJF. Editorial: Clonal hematopoiesis: a link between inflammation, ageing and cardiovascular disease. Curr Opin Lipidol 2020; 31:102-103. [PMID: 32132414 DOI: 10.1097/mol.0000000000000661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Uwe J F Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
303
|
Yki-Järvinen H. Ceramides: A Cause of Insulin Resistance in Nonalcoholic Fatty Liver Disease in Both Murine Models and Humans. Hepatology 2020; 71:1499-1501. [PMID: 31899812 DOI: 10.1002/hep.31095] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland
| |
Collapse
|
304
|
Hypercholesterolemia risk-associated GPR146 is an orphan G-protein coupled receptor that regulates blood cholesterol levels in humans and mice. Cell Res 2020; 30:363-365. [PMID: 32203133 DOI: 10.1038/s41422-020-0303-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/05/2020] [Indexed: 12/24/2022] Open
|
305
|
Abstract
Nonalcoholic fatty liver disease is strongly associated with obesity and the metabolic syndrome, but genetic factors also contribute to disease susceptibility. Human genetic studies have identified several common genetic variants contributing to nonalcoholic fatty liver disease initiation and progression. These findings have provided new insights into the pathogenesis of nonalcoholic fatty liver disease and opened up new avenues for the development of therapeutic interventions. In this review, we summarize the current state of knowledge about the genetic determinants of nonalcoholic fatty liver disease, focusing on the most robustly validated genetic risk factors and on recently discovered modifiers of disease progression.
Collapse
Affiliation(s)
- Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8591, USA.
| |
Collapse
|
306
|
Hu Y, Graff M, Haessler J, Buyske S, Bien SA, Tao R, Highland HM, Nishimura KK, Zubair N, Lu Y, Verbanck M, Hilliard AT, Klarin D, Damrauer SM, Ho YL, the VA Million Veteran Program, Wilson PWF, Chang KM, Tsao PS, Cho K, O’Donnell CJ, Assimes TL, Petty LE, Below JE, Dikilitas O, Schaid DJ, Kosel ML, Kullo IJ, Rasmussen-Torvik LJ, Jarvik GP, Feng Q, Wei WQ, Larson EB, Mentch FD, Almoguera B, Sleiman PM, Raffield LM, Correa A, Martin LW, Daviglus M, Matise TC, Ambite JL, Carlson CS, Do R, Loos RJF, Wilkens LR, Le Marchand L, Haiman C, Stram DO, Hindorff LA, North KE, Kooperberg C, Cheng I, Peters U. Minority-centric meta-analyses of blood lipid levels identify novel loci in the Population Architecture using Genomics and Epidemiology (PAGE) study. PLoS Genet 2020; 16:e1008684. [PMID: 32226016 PMCID: PMC7145272 DOI: 10.1371/journal.pgen.1008684] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 04/09/2020] [Accepted: 02/19/2020] [Indexed: 11/18/2022] Open
Abstract
Lipid levels are important markers for the development of cardio-metabolic diseases. Although hundreds of associated loci have been identified through genetic association studies, the contribution of genetic factors to variation in lipids is not fully understood, particularly in U.S. minority groups. We performed genome-wide association analyses for four lipid traits in over 45,000 ancestrally diverse participants from the Population Architecture using Genomics and Epidemiology (PAGE) Study, followed by a meta-analysis with several European ancestry studies. We identified nine novel lipid loci, five of which showed evidence of replication in independent studies. Furthermore, we discovered one novel gene in a PrediXcan analysis, minority-specific independent signals at eight previously reported loci, and potential functional variants at two known loci through fine-mapping. Systematic examination of known lipid loci revealed smaller effect estimates in African American and Hispanic ancestry populations than those in Europeans, and better performance of polygenic risk scores based on minority-specific effect estimates. Our findings provide new insight into the genetic architecture of lipid traits and highlight the importance of conducting genetic studies in diverse populations in the era of precision medicine.
Collapse
Affiliation(s)
- Yao Hu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jeffrey Haessler
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Steven Buyske
- Department of Statistics and Biostatistics, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Stephanie A. Bien
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- The Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Heather M. Highland
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Katherine K. Nishimura
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Niha Zubair
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Marie Verbanck
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Austin T. Hilliard
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Derek Klarin
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Boston VA Healthcare System, Boston, Massachusetts, United States of America
| | - Scott M. Damrauer
- Emory Clinical Cardiovascular Research Institute, Atlanta, Georgia, United States of America
- Corporal Michael Crescenz VA Medical Center, Philadelphia, Pennsylvania, United States of America
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Yuk-Lam Ho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, United States of America
| | | | - Peter W. F. Wilson
- Emory Clinical Cardiovascular Research Institute, Atlanta, Georgia, United States of America
- Atlanta VA Medical Center, Decatur, Georgia, United States of America
| | - Kyong-Mi Chang
- Corporal Michael Crescenz VA Medical Center, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Philip S. Tsao
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, United States of America
| | - Christopher J. O’Donnell
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Themistocles L. Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Lauren E. Petty
- The Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Epidemiology, Human Genetics & Environmental Sciences, University of Texas School of Public Health, Houston, Texas, United States of America
| | - Jennifer E. Below
- The Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Epidemiology, Human Genetics & Environmental Sciences, University of Texas School of Public Health, Houston, Texas, United States of America
| | - Ozan Dikilitas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Daniel J. Schaid
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Matthew L. Kosel
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Iftikhar J. Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Laura J. Rasmussen-Torvik
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Gail P. Jarvik
- Department of Medicine, University of Washington Medical Center, Seattle, Washington, United States of America
| | - Qiping Feng
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Wei-Qi Wei
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Eric B. Larson
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington, United States of America
| | - Frank D. Mentch
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Berta Almoguera
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Patrick M. Sleiman
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Adolfo Correa
- Departments of Medicine, Pediatrics, and Population Health Science, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Lisa W. Martin
- School of Medicine and Health Sciences, George Washington University, Washington, District of Columbia, United States of America
| | - Martha Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Tara C. Matise
- Department of Statistics and Biostatistics, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Jose Luis Ambite
- Information Sciences Institute, University of Southern California, Marina del Rey, California, United States of America
| | - Christopher S. Carlson
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lynne R. Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Chris Haiman
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Daniel O. Stram
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Lucia A. Hindorff
- Division of Genomic Medicine, NIH National Human Genome Research Institute, Bethesda, Maryland, United States of America
| | - Kari E. North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Iona Cheng
- Cancer Prevention Institute of California, Fremont, California, United States of America
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| |
Collapse
|
307
|
Riveros-Mckay F, Oliver-Williams C, Karthikeyan S, Walter K, Kundu K, Ouwehand WH, Roberts D, Di Angelantonio E, Soranzo N, Danesh J, INTERVAL Study, Wheeler E, Zeggini E, Butterworth AS, Barroso I. The influence of rare variants in circulating metabolic biomarkers. PLoS Genet 2020; 16:e1008605. [PMID: 32150548 PMCID: PMC7108731 DOI: 10.1371/journal.pgen.1008605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 03/31/2020] [Accepted: 01/10/2020] [Indexed: 12/19/2022] Open
Abstract
Circulating metabolite levels are biomarkers for cardiovascular disease (CVD). Here we studied, association of rare variants and 226 serum lipoproteins, lipids and amino acids in 7,142 (discovery plus follow-up) healthy participants. We leveraged the information from multiple metabolite measurements on the same participants to improve discovery in rare variant association analyses for gene-based and gene-set tests by incorporating correlated metabolites as covariates in the validation stage. Gene-based analysis corrected for the effective number of tests performed, confirmed established associations at APOB, APOC3, PAH, HAL and PCSK (p<1.32x10-7) and identified novel gene-trait associations at a lower stringency threshold with ACSL1, MYCN, FBXO36 and B4GALNT3 (p<2.5x10-6). Regulation of the pyruvate dehydrogenase (PDH) complex was associated for the first time, in gene-set analyses also corrected for effective number of tests, with IDL and LDL parameters, as well as circulating cholesterol (pMETASKAT<2.41x10-6). In conclusion, using an approach that leverages metabolite measurements obtained in the same participants, we identified novel loci and pathways involved in the regulation of these important metabolic biomarkers. As large-scale biobanks continue to amass sequencing and phenotypic information, analytical approaches such as ours will be useful to fully exploit the copious amounts of biological data generated in these efforts.
Collapse
Affiliation(s)
| | - Clare Oliver-Williams
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- Homerton College, Cambridge, United Kingdom
| | - Savita Karthikeyan
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | | | - Kousik Kundu
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Willem H. Ouwehand
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - David Roberts
- The National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- NHS Blood and Transplant—Oxford Centre, Level 2, John Radcliffe Hospital, Oxford, United Kingdom
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Emanuele Di Angelantonio
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- The National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
| | - Nicole Soranzo
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - John Danesh
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- The National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
| | | | - Eleanor Wheeler
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Eleftheria Zeggini
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Institute of Translational Genomics, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Adam S. Butterworth
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- The National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
| | - Inês Barroso
- Wellcome Sanger Institute, Cambridge, United Kingdom
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| |
Collapse
|
308
|
Amorós-Pérez M, Fuster JJ. Clonal hematopoiesis driven by somatic mutations: A new player in atherosclerotic cardiovascular disease. Atherosclerosis 2020; 297:120-126. [PMID: 32109665 DOI: 10.1016/j.atherosclerosis.2020.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023]
Abstract
The accumulation of acquired mutations is an inevitable consequence of the aging process, but its pathophysiological relevance has remained largely unexplored beyond cancer. Most of these mutations have little or no functional consequences, but in a few rare instances, a mutation may arise that confers a competitive advantage to a stem cell, leading to its clonal expansion. When such a mutation occurs in hematopoietic stem cells, it leads to a situation of clonal hematopoiesis, which has the potential to affect multiple tissues beyond the bone marrow, as the clonal expansion of the mutant stem cell is extended to circulating blood cells and tissue-infiltrating immune cells. Recent genomics and experimental studies have provided support to the notion that this somatic mutation-driven clonal hematopoiesis contributes to vascular inflammation and the development of atherosclerosis and related cardiovascular and cerebrovascular ischemic events. Here, we review our current understanding of this emerging cardiovascular risk modifier and the mechanisms underlying its connection to atherosclerosis development.
Collapse
Affiliation(s)
- Marta Amorós-Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - José J Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
309
|
Abstract
With regard to heritability of phenotypes, the serum triglyceride level is considered to be highly heritable, with approximately 50% of its variability estimated to derive from parents. Thus, approximately 50% could be modifiable via environmental factors, including lifestyle and medications. Lipoproteins are definitive risk factors for atherosclerotic cardiovascular disease (ASCVD); among these, low-density lipoprotein (LDL) particles have been established as a causal factor for the development of ASCVD. Recently, triglyceride-rich lipoproteins have emerged as additional lipoproteins, which should be considered as residual targets for ASCVD risk reduction by LDL-lowering therapies. Compared with LDL particles, triglyceride-rich lipoproteins are significantly increased in the postprandial state, making it difficult to assess their clinical relevance. However, numerous pieces of evidence suggest that fasting and non-fasting triglycerides are associated with ASCVD. In addition, a recent meta-analysis of a Mendelian randomization study suggests that consideration of apolipoprotein B (APOB) might be better than considering LDL and triglyceride-rich lipoproteins separately. In this review, we examine (1) how triglyceride levels are determined by genetics, (2) lessons from extreme cases exhibiting severe hypertriglyceridemia, and (3) why triglycerides are important, by highlighting clinical and genetic evidence of their associations with ASCVD risk.
Collapse
Affiliation(s)
- Hayato Tada
- Department of Cardiology, Kanazawa University, Graduate School of Medical Sciences, Kanazawa, Japan.
| | - Masayuki Takamura
- Department of Cardiology, Kanazawa University, Graduate School of Medical Sciences, Kanazawa, Japan
| | - Masa-Aki Kawashiri
- Department of Cardiology, Kanazawa University, Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
310
|
Reggiani F, Carraro M, Belligoli A, Sanna M, dal Prà C, Favaretto F, Ferrari C, Vettor R, Tosatto SCE. In silico prediction of blood cholesterol levels from genotype data. PLoS One 2020; 15:e0227191. [PMID: 32040480 PMCID: PMC7010235 DOI: 10.1371/journal.pone.0227191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/14/2019] [Indexed: 11/18/2022] Open
Abstract
In this work we present a framework for blood cholesterol levels prediction from genotype data. The predictor is based on an algorithm for cholesterol metabolism simulation available in literature, implemented and optimized by our group in the R language. The main weakness of the former simulation algorithm was the need of experimental data to simulate mutations in genes altering the cholesterol metabolism. This caveat strongly limited the application of the model in the clinical practice. In this work we present how this limitation could be bypassed thanks to an optimization of model parameters based on patient cholesterol levels retrieved from literature. Prediction performance has been assessed taking into consideration several scoring indices currently used for performance evaluation of machine learning methods. Our assessment shows how the optimization phase improved model performance, compared to the original version available in literature.
Collapse
Affiliation(s)
- Francesco Reggiani
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Marco Carraro
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Anna Belligoli
- Clinica Medica 3, Department of Medicine—DIMED, School of Medicine, University of Padua, Padua, Italy
| | - Marta Sanna
- Clinica Medica 3, Department of Medicine—DIMED, School of Medicine, University of Padua, Padua, Italy
| | - Chiara dal Prà
- Clinica Medica 3, Department of Medicine—DIMED, School of Medicine, University of Padua, Padua, Italy
| | - Francesca Favaretto
- Clinica Medica 3, Department of Medicine—DIMED, School of Medicine, University of Padua, Padua, Italy
| | - Carlo Ferrari
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Roberto Vettor
- Clinica Medica 3, Department of Medicine—DIMED, School of Medicine, University of Padua, Padua, Italy
| | - Silvio C. E. Tosatto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- CNR Institute of Neuroscience, Padua, Italy
- * E-mail:
| |
Collapse
|
311
|
Brouwers MCGJ, Simons N, Stehouwer CDA, Isaacs A. Non-alcoholic fatty liver disease and cardiovascular disease: assessing the evidence for causality. Diabetologia 2020; 63:253-260. [PMID: 31713012 PMCID: PMC6946734 DOI: 10.1007/s00125-019-05024-3] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is highly prevalent among individuals with type 2 diabetes. Although epidemiological studies have shown that NAFLD is associated with cardiovascular disease (CVD), it remains unknown whether NAFLD is an active contributor or an innocent bystander. Plasma lipids, low-grade inflammation, impaired fibrinolysis and hepatokines are potential mediators of the relationship between NAFLD and CVD. The Mendelian randomisation approach can help to make causal inferences. Studies that used common variants in PNPLA3, TM6SF2 and GCKR as instruments to investigate the relationship between NAFLD and coronary artery disease (CAD) have reported contrasting results. Variants in PNPLA3 and TM6SF2 were found to protect against CAD, whereas variants in GCKR were positively associated with CAD. Since all three genes have been associated with non-alcoholic steatohepatitis, the second stage of NAFLD, the question of whether low-grade inflammation is an important mediator of the relationship between NAFLD and CAD arises. In contrast, the differential effects of these genes on plasma lipids (i.e. lipid-lowering for PNPLA3 and TM6SF2, and lipid-raising for GCKR) strongly suggest that plasma lipids account for their differential effects on CAD risk. This concept has recently been confirmed in an extended set of 12 NAFLD susceptibility genes. From these studies it appears that plasma lipids are an important mediator between NAFLD and CVD risk. These findings have important clinical implications, particularly for the design of anti-NAFLD drugs that also affect lipid metabolism.
Collapse
Affiliation(s)
- Martijn C G J Brouwers
- Division of Endocrinology and Metabolic Disease, Department of Internal Medicine, Maastricht University Medical Centre, P. Debyelaan 25, 6229 HX, Maastricht, the Netherlands.
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.
| | - Nynke Simons
- Division of Endocrinology and Metabolic Disease, Department of Internal Medicine, Maastricht University Medical Centre, P. Debyelaan 25, 6229 HX, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Coen D A Stehouwer
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Aaron Isaacs
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
- Department of Biochemistry, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
312
|
Loaiza N, Hartgers ML, Reeskamp LF, Balder JW, Rimbert A, Bazioti V, Wolters JC, Winkelmeijer M, Jansen HPG, Dallinga-Thie GM, Volta A, Huijkman N, Smit M, Kloosterhuis N, Koster M, Svendsen AF, van de Sluis B, Hovingh GK, Grefhorst A, Kuivenhoven JA. Taking One Step Back in Familial Hypercholesterolemia: STAP1 Does Not Alter Plasma LDL (Low-Density Lipoprotein) Cholesterol in Mice and Humans. Arterioscler Thromb Vasc Biol 2020; 40:973-985. [PMID: 31996024 PMCID: PMC7098433 DOI: 10.1161/atvbaha.119.313470] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE STAP1, encoding for STAP1 (signal transducing adaptor family member 1), has been reported as a candidate gene associated with familial hypercholesterolemia. Unlike established familial hypercholesterolemia genes, expression of STAP1 is absent in liver but mainly observed in immune cells. In this study, we set out to validate STAP1 as a familial hypercholesterolemia gene. Approach and Results: A whole-body Stap1 knockout mouse model (Stap1-/-) was generated and characterized, without showing changes in plasma lipid levels compared with controls. In follow-up studies, bone marrow from Stap1-/- mice was transplanted to Ldlr-/- mice, which did not show significant changes in plasma lipid levels or atherosclerotic lesions. To functionally assess whether STAP1 expression in B cells can affect hepatic function, HepG2 cells were cocultured with peripheral blood mononuclear cells isolated from heterozygotes carriers of STAP1 variants and controls. The peripheral blood mononuclear cells from STAP1 variant carriers and controls showed similar LDLR mRNA and protein levels. Also, LDL (low-density lipoprotein) uptake by HepG2 cells did not differ upon coculturing with peripheral blood mononuclear cells isolated from either STAP1 variant carriers or controls. In addition, plasma lipid profiles of 39 carriers and 71 family controls showed no differences in plasma LDL cholesterol, HDL (high-density lipoprotein) cholesterol, triglycerides, and lipoprotein(a) levels. Similarly, B-cell populations did not differ in a group of 10 STAP1 variant carriers and 10 age- and sex-matched controls. Furthermore, recent data from the UK Biobank do not show association between STAP1 rare gene variants and LDL cholesterol. CONCLUSIONS Our combined studies in mouse models and carriers of STAP1 variants indicate that STAP1 is not a familial hypercholesterolemia gene.
Collapse
Affiliation(s)
- Natalia Loaiza
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Merel L Hartgers
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, the Netherlands (M.L.H., L.F.R., G.M.D.-T., G.K.H.)
| | - Laurens F Reeskamp
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, the Netherlands (M.L.H., L.F.R., G.M.D.-T., G.K.H.)
| | - Jan-Willem Balder
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands.,Department of Vascular Medicine (J.-W.B.), University Medical Center Groningen, University of Groningen, the Netherlands.,Department of Cardiology, University Medical Center Utrecht, the Netherlands (J.-W.B.)
| | - Antoine Rimbert
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands.,L'institut du thorax, INSERM, CNRS, Université de Nantes, France (A.R.)
| | - Venetia Bazioti
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Justina C Wolters
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Maaike Winkelmeijer
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, the Netherlands (M.W., H.P.G.J., A.G.)
| | - Hans P G Jansen
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, the Netherlands (M.W., H.P.G.J., A.G.)
| | - Geesje M Dallinga-Thie
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, the Netherlands (M.L.H., L.F.R., G.M.D.-T., G.K.H.)
| | - Andrea Volta
- Department of Experimental and Clinical Medicine, University of Florence, Italy (A.V.)
| | - Nicolette Huijkman
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Marieke Smit
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Niels Kloosterhuis
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Mirjam Koster
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Arthur F Svendsen
- Laboratory of Ageing Biology and Stem Cells, European Institute for the Biology of Aging (ERIBA) (A.F.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Bart van de Sluis
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands.,iPSC/CRISPR Center Groningen (B.v.d.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - G Kees Hovingh
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, the Netherlands (M.L.H., L.F.R., G.M.D.-T., G.K.H.)
| | - Aldo Grefhorst
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, the Netherlands (M.W., H.P.G.J., A.G.)
| | - Jan Albert Kuivenhoven
- From the Department of Pediatrics, Molecular Genetics Section (N.L., J.-W.B., A.R., V.B., J.C.W., N.H., M.S., N.K., M.K., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
313
|
Larsen LE, Stoekenbroek RM, Kastelein JJP, Holleboom AG. Moving Targets: Recent Advances in Lipid-Lowering Therapies. Arterioscler Thromb Vasc Biol 2020; 39:349-359. [PMID: 30676072 DOI: 10.1161/atvbaha.118.312028] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Statin therapy has delivered tremendous value to society by improving the burden of atherosclerotic cardiovascular disease. Nonetheless, atherosclerotic cardiovascular disease remains the leading cause of death globally. Technological advances such as in the field of genomics have revolutionized drug discovery and development and have revealed novel therapeutic targets to lower low-density lipoprotein cholesterol (LDL-C), as well as other detrimental lipids and lipoproteins. Therapeutic LDL-C lowering prevents atherosclerotic cardiovascular disease with an effect size proportional to absolute LDL-C reductions and time of exposure. This understanding supports the notion that reducing cumulative LDL-C exposure should be a key therapeutic target. PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibiting monoclonal antibodies provides the possibility of reducing LDL-C to very low levels. Novel therapeutic platforms such as RNA inhibition present opportunities to combine robust lipid lowering with infrequent dosing regimens, introducing therapies with vaccine-like properties. The position of lipid-lowering therapies with targets other than LDL-C, such as Lp(a) [lipoprotein(a)], TRL (triglyceride-rich lipoproteins), and remnant cholesterol, will likely be determined by the results of ongoing clinical trials. Current evidence suggests that reducing Lp(a) or TRLs could attenuate atherosclerotic cardiovascular disease risk in specific categories of patients. This review provides an overview of the latest therapeutic developments, focusing on their mechanisms, efficacy, and safety.
Collapse
Affiliation(s)
- Lars E Larsen
- From the Department of Vascular Medicine, Amsterdam University Medical Center, The Netherlands
| | - Robert M Stoekenbroek
- From the Department of Vascular Medicine, Amsterdam University Medical Center, The Netherlands
| | - John J P Kastelein
- From the Department of Vascular Medicine, Amsterdam University Medical Center, The Netherlands
| | - Adriaan G Holleboom
- From the Department of Vascular Medicine, Amsterdam University Medical Center, The Netherlands
| |
Collapse
|
314
|
Butler AA, Graham JL, Stanhope KL, Wong S, King S, Bremer AA, Krauss RM, Hamilton J, Havel PJ. Role of angiopoietin-like protein 3 in sugar-induced dyslipidemia in rhesus macaques: suppression by fish oil or RNAi. J Lipid Res 2020; 61:376-386. [PMID: 31919051 DOI: 10.1194/jlr.ra119000423] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Angiopoietin-like protein 3 (ANGPTL3) inhibits lipid clearance and is a promising target for managing cardiovascular disease. Here we investigated the effects of a high-sugar (high-fructose) diet on circulating ANGPTL3 concentrations in rhesus macaques. Plasma ANGPTL3 concentrations increased ∼30% to 40% after 1 and 3 months of a high-fructose diet (both P < 0.001 vs. baseline). During fructose-induced metabolic dysregulation, plasma ANGPTL3 concentrations were positively correlated with circulating indices of insulin resistance [assessed with fasting insulin and the homeostatic model assessment of insulin resistance (HOMA-IR)], hypertriglyceridemia, adiposity (assessed as leptin), and systemic inflammation [C-reactive peptide (CRP)] and negatively correlated with plasma levels of the insulin-sensitizing hormone adropin. Multiple regression analyses identified a strong association between circulating APOC3 and ANGPTL3 concentrations. Higher baseline plasma levels of both ANGPTL3 and APOC3 were associated with an increased risk for fructose-induced insulin resistance. Fish oil previously shown to prevent insulin resistance and hypertriglyceridemia in this model prevented increases of ANGPTL3 without affecting systemic inflammation (increased plasma CRP and interleukin-6 concentrations). ANGPTL3 RNAi lowered plasma concentrations of ANGPTL3, triglycerides (TGs), VLDL-C, APOC3, and APOE. These decreases were consistent with a reduced risk of atherosclerosis. In summary, dietary sugar-induced increases of circulating ANGPTL3 concentrations after metabolic dysregulation correlated positively with leptin levels, HOMA-IR, and dyslipidemia. Targeting ANGPTL3 expression with RNAi inhibited dyslipidemia by lowering plasma TGs, VLDL-C, APOC3, and APOE levels in rhesus macaques.
Collapse
Affiliation(s)
- Andrew A Butler
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| | - James L Graham
- Department of Molecular Biosciences, University of California, Davis, Davis, CA.,School of Veterinary Medicine, California National Primate Research Center, and Department of Nutrition, University of California, Davis, Davis, CA
| | - Kimber L Stanhope
- Department of Molecular Biosciences, University of California, Davis, Davis, CA.,School of Veterinary Medicine, California National Primate Research Center, and Department of Nutrition, University of California, Davis, Davis, CA
| | - So Wong
- Arrowhead Pharmaceuticals, Pasadena, CA
| | - Sarah King
- Children's Hospital Oakland Research Institute, Oakland, CA
| | - Andrew A Bremer
- Department of Pediatrics, Vanderbilt University, Nashville, TN
| | | | | | - Peter J Havel
- Department of Molecular Biosciences, University of California, Davis, Davis, CA .,School of Veterinary Medicine, California National Primate Research Center, and Department of Nutrition, University of California, Davis, Davis, CA
| |
Collapse
|
315
|
Liu X, Zhang Y, Liang H, Xu Y. Overexpression of microRNA-216a-3p Accelerates the Inflammatory Response in Cardiomyocytes in Type 2 Diabetes Mellitus by Targeting IFN-α2. Front Endocrinol (Lausanne) 2020; 11:522340. [PMID: 33329376 PMCID: PMC7729074 DOI: 10.3389/fendo.2020.522340] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 09/03/2020] [Indexed: 11/22/2022] Open
Abstract
Background: Type 2 diabetes mellitus (T2DM) is a chronic, hyperglycemia-associated, metabolic disorder. Heart disease is a major complication of T2DM. The present study aimed to explore the effects of miR-216a-3p on cardiomyocyte proliferation, apoptosis, and inflammation in T2DM through the Toll-like receptor (TLR) pathway involving interferon-α2 (IFN-α2) mediation. Methods: T2DM was induced in rats by a high-fat diet, in combination with an intraperitoneal injection of low-dose streptozotocin. ELISAs were conducted to measure inflammatory-related factors in serum. Next, isolated cardiomyocytes were used in loss- and gain-of-function experiments, followed by MTT and flow cytometry assays, conducted to evaluate cell proliferation, cell cycle, and apoptosis. Results: Our results revealed an increase in the inflammatory response in T2DM rat models, accompanied by significantly increased expression of miR-216a-3p and TLR pathway-related genes. However, a decrease in the expression of IFN-α2 was observed. Moreover, the presence of an miR-216a-3p inhibitor and si-IFN-α2 increased the expression of TLR pathway-related genes and cell apoptosis, whereas cell proliferation was significantly decreased in the cardiomyocytes. Conclusion: We found that in T2DM, miR-216a-3p inhibited the proliferation and enhanced the apoptosis of cardiomyocytes and generated an inflammatory response through activation of the TLR pathway and targeting of IFN-α2.
Collapse
Affiliation(s)
- Xiaomeng Liu
- The 2nd Ward, Department of Endocrinology and Metabolism, Linyi People's Hospital, Linyi, China
| | - Yusong Zhang
- Imaging Center, Linyi People's Hospital, Linyi, China
| | - Hongwei Liang
- Department of Health Care, Linyi People's Hospital, Linyi, China
| | - Yanchao Xu
- The 2nd Ward, Department of Endocrinology and Metabolism, Linyi People's Hospital, Linyi, China
- *Correspondence: Yanchao Xu
| |
Collapse
|
316
|
Eslam M, George J. Genetic contributions to NAFLD: leveraging shared genetics to uncover systems biology. Nat Rev Gastroenterol Hepatol 2020; 17:40-52. [PMID: 31641249 DOI: 10.1038/s41575-019-0212-0] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2019] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects around a quarter of the global population, paralleling worldwide increases in obesity and metabolic syndrome. NAFLD arises in the context of systemic metabolic dysfunction that concomitantly amplifies the risk of cardiovascular disease and diabetes. These interrelated conditions have long been recognized to have a heritable component, and advances using unbiased association studies followed by functional characterization have created a paradigm for unravelling the genetic architecture of these conditions. A novel perspective is to characterize the shared genetic basis of NAFLD and other related disorders. This information on shared genetic risks and their biological overlap should in future enable the development of precision medicine approaches through better patient stratification, and enable the identification of preventive and therapeutic strategies. In this Review, we discuss current knowledge of the genetic basis of NAFLD and of possible pleiotropy between NAFLD and other liver diseases as well as other related metabolic disorders. We also discuss evidence of causality in NAFLD and other related diseases and the translational significance of such evidence, and future challenges from the study of genetic pleiotropy.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia.
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
317
|
Yang T, Kim J, Wu C, Ma Y, Wei P, Pan W. An adaptive test for meta-analysis of rare variant association studies. Genet Epidemiol 2020; 44:104-116. [PMID: 31830326 PMCID: PMC6980317 DOI: 10.1002/gepi.22273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/12/2019] [Accepted: 11/25/2019] [Indexed: 01/02/2023]
Abstract
Single genome-wide studies may be underpowered to detect trait-associated rare variants with moderate or weak effect sizes. As a viable alternative, meta-analysis is widely used to increase power by combining different studies. The power of meta-analysis critically depends on the underlying association patterns and heterogeneity levels, which are unknown and vary from locus to locus. However, existing methods mainly focus on one or only a few combinations of the association pattern and heterogeneity level, thus may lose power in many situations. To address this issue, we propose a general and unified framework by combining a class of tests including and beyond some existing ones, leading to high power across a wide range of scenarios. We demonstrate that the proposed test is more powerful than some existing methods in simulation studies, then show their performance with the NHLBI Exome-Sequencing Project (ESP) data. One gene (B4GALNT2) was found by our proposed test, but not by others, to be statistically significantly associated with plasma triglyceride. The signal was driven by African-ancestry subjects but it was previously reported to be associated with coronary artery disease among European-ancestry subjects. We implemented our method in an R package aSPUmeta, publicly available at https://github.com/ytzhong/metaRV and will be on CRAN soon.
Collapse
Affiliation(s)
- Tianzhong Yang
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Junghi Kim
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Chong Wu
- Department of Statistics, Florida State University, Tallahassee, FL, USA
| | - Yiding Ma
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Pan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
318
|
Borén J, Packard CJ, Taskinen MR. The Roles of ApoC-III on the Metabolism of Triglyceride-Rich Lipoproteins in Humans. Front Endocrinol (Lausanne) 2020; 11:474. [PMID: 32849270 PMCID: PMC7399058 DOI: 10.3389/fendo.2020.00474] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. It is well-established based on evidence accrued during the last three decades that high plasma concentrations of cholesterol-rich atherogenic lipoproteins are causatively linked to CVD, and that lowering these reduces atherosclerotic cardiovascular events in humans (1-9). Historically, most attention has been on low-density lipoproteins (LDL) since these are the most abundant atherogenic lipoproteins in the circulation, and thus the main carrier of cholesterol into the artery wall. However, with the rise of obesity and insulin resistance in many populations, there is increasing interest in the role of triglyceride-rich lipoproteins (TRLs) and their metabolic remnants, with accumulating evidence showing they too are causatively linked to CVD. Plasma triglyceride, measured either in the fasting or non-fasting state, is a useful index of the abundance of TRLs and recent research into the biology and genetics of triglyceride heritability has provided new insight into the causal relationship of TRLs with CVD. Of the genetic factors known to influence plasma triglyceride levels variation in APOC3- the gene for apolipoprotein (apo) C-III - has emerged as being particularly important as a regulator of triglyceride transport and a novel therapeutic target to reduce dyslipidaemia and CVD risk (10).
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- *Correspondence: Jan Borén
| | - Chris J. Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Marja-Riitta Taskinen
- Research Programs Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| |
Collapse
|
319
|
Williams DM, Finan C, Schmidt AF, Burgess S, Hingorani AD. Lipid lowering and Alzheimer disease risk: A mendelian randomization study. Ann Neurol 2020; 87:30-39. [PMID: 31714636 PMCID: PMC6944510 DOI: 10.1002/ana.25642] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To examine whether genetic variation affecting the expression or function of lipid-lowering drug targets is associated with Alzheimer disease (AD) risk, to evaluate the potential impact of long-term exposure to corresponding therapeutics. METHODS We conducted Mendelian randomization analyses using variants in genes that encode the protein targets of several approved lipid-lowering drug classes: HMGCR (encoding the target for statins), PCSK9 (encoding the target for PCSK9 inhibitors, eg, evolocumab and alirocumab), NPC1L1 (encoding the target for ezetimibe), and APOB (encoding the target of mipomersen). Variants were weighted by associations with low-density lipoprotein cholesterol (LDL-C) using data from lipid genetics consortia (n up to 295,826). We meta-analyzed Mendelian randomization estimates for regional variants weighted by LDL-C on AD risk from 2 large samples (total n = 24,718 cases, 56,685 controls). RESULTS Models for HMGCR, APOB, and NPC1L1 did not suggest that the use of related lipid-lowering drug classes would affect AD risk. In contrast, genetically instrumented exposure to PCSK9 inhibitors was predicted to increase AD risk in both of the AD samples (combined odds ratio per standard deviation lower LDL-C inducible by the drug target = 1.45, 95% confidence interval = 1.23-1.69). This risk increase was opposite to, although more modest than, the degree of protection from coronary artery disease predicted by these same methods for PCSK9 inhibition. INTERPRETATION We did not identify genetic support for the repurposing of statins, ezetimibe, or mipomersen for AD prevention. Notwithstanding caveats to this genetic evidence, pharmacovigilance for AD risk among users of PCSK9 inhibitors may be warranted. ANN NEUROL 2020;87:30-39.
Collapse
Affiliation(s)
- Dylan M. Williams
- Medical Research Council Unit for Lifelong Health and Ageing at University College LondonUniversity College LondonLondonUnited Kingdom
- Department of Medical Epidemiology & BiostatisticsKarolinska InstituteStockholmSweden
| | - Chris Finan
- Institute of Cardiovascular Science, Faculty of Population HealthUniversity College LondonLondonUnited Kingdom
- Health Data Research UKLondonUnited Kingdom
- British Heart Foundation University College London Research AcceleratorLondonUnited Kingdom
| | - Amand F. Schmidt
- Institute of Cardiovascular Science, Faculty of Population HealthUniversity College LondonLondonUnited Kingdom
- Health Data Research UKLondonUnited Kingdom
- British Heart Foundation University College London Research AcceleratorLondonUnited Kingdom
- Department of Cardiology, Division Heart and LungsUniversity Medical Center UtrechtUtrechtthe Netherlands
| | - Stephen Burgess
- Medical Research Council Biostatistics UnitUniversity of CambridgeCambridgeUnited Kingdom
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary CareUniversity of CambridgeCambridgeUnited Kingdom
| | - Aroon D. Hingorani
- Institute of Cardiovascular Science, Faculty of Population HealthUniversity College LondonLondonUnited Kingdom
- Health Data Research UKLondonUnited Kingdom
- British Heart Foundation University College London Research AcceleratorLondonUnited Kingdom
| |
Collapse
|
320
|
Chen L, Yao Y, Jin C, Wu S, Liu Q, Li J, Ma Y, Xu Y, Zhong Y. Integrative genomic analysis identified common regulatory networks underlying the correlation between coronary artery disease and plasma lipid levels. BMC Cardiovasc Disord 2019; 19:310. [PMID: 31870308 PMCID: PMC6927120 DOI: 10.1186/s12872-019-01271-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/21/2019] [Indexed: 12/15/2022] Open
Abstract
Background Coronary artery disease (CAD) and plasma lipid levels are highly correlated, indicating the presence of common pathways between them. Nevertheless, the molecular pathways underlying the pathogenic comorbidities for both traits remain poorly studied. We sought to identify common pathways and key driver genes by performing a comprehensive integrative analysis based on multi-omic datasets. Methods By performing a pathway-based analysis of GWAS summary data, we identified that lipoprotein metabolism process-related pathways were significantly associated with CAD risk. Based on LD score regression analysis of CAD-related SNPs, significant heritability enrichments were observed in the cardiovascular and digestive system, as well as in liver and gastrointestinal tissues, which are the main regulators for lipid level. Results We found there existed significant genetic correlation between CAD and other lipid metabolism related traits (the smallest P value < 1 × 10− 16). A total of 13 genes (e.g., LPA, APOC1, APOE and SLC22A3) was found to be overlapped between CAD and plasma lipid levels. By using the data-driven approach that integrated transcriptome information, we discovered co-expression modules associated prominently with both CAD and plasma lipids. With the detailed topology information on gene-gene regulatory relationship, we illustrated that the identified hub genes played important roles in the pathogenesis of CAD and plasma lipid turbulence. Conclusion Together, we identified the shared molecular mechanisms underlying the correlation between CAD and plasma lipid levels.
Collapse
Affiliation(s)
- Liuying Chen
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Yinghao Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Shen Wu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunlong Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yizhou Xu
- Zhejiang Chinese Medical University, Hangzhou, China.,Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yigang Zhong
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
321
|
Affiliation(s)
- Andrés Hidalgo
- From the Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.H.)
| | - Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (A.R.T.)
| |
Collapse
|
322
|
Smit RAJ, Trompet S, Leong A, Goodarzi MO, Postmus I, Warren H, Theusch E, Barnes MR, Arsenault BJ, Li X, Feng Q, Chasman DI, Cupples LA, Hitman GA, Krauss RM, Psaty BM, Rotter JI, Cessie SL, Stein CM, Jukema JW. Statin-induced LDL cholesterol response and type 2 diabetes: a bidirectional two-sample Mendelian randomization study. THE PHARMACOGENOMICS JOURNAL 2019; 20:462-470. [PMID: 31801993 PMCID: PMC7260089 DOI: 10.1038/s41397-019-0125-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 11/11/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023]
Abstract
It remains unclear whether the increased risk of new-onset type 2 diabetes (T2D) seen in statin users is due to low LDL-C concentrations, or due to the statin-induced proportional change in LDL-C. In addition, genetic instruments have not been proposed before to examine whether liability to T2D might cause greater proportional statin-induced LDL-C lowering. Using summary level statistics from the Genomic Investigation of Statin Therapy (GIST, nmax=40,914) and DIAGRAM (nmax=159,208) consortia, we found a positive genetic correlation between LDL-C statin response and T2D using LD score regression (rgenetic=0.36, s.e.=0.13). However, mendelian randomization analyses did not provide support for statin response having a causal effect on T2D risk (OR 1.00 (95%CI: 0.97, 1.03) per 10% increase in statin response), nor that liability to T2D has a causal effect on statin-induced LDL-C response (0.20% increase in response (95%CI: −0.40, 0.80) per doubling of odds of liability to T2D). Although we found no evidence to suggest that proportional statin response influences T2D risk, a definitive assessment should be made in populations comprised exclusively of statin-users, as the presence of non-statin users in the DIAGRAM dataset may have substantially diluted our effect estimate.
Collapse
Affiliation(s)
- Roelof A J Smit
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands. .,Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Aaron Leong
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA.,Harvard Medical School, Boston, MA, 02115, USA
| | - Mark O Goodarzi
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Iris Postmus
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Helen Warren
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK.,National Institute for Health Research, Barts Cardiovascular Biomedical Research Center, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Elizabeth Theusch
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr Way, Oakland, CA, 94609, USA
| | - Michael R Barnes
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK.,National Institute for Health Research, Barts Cardiovascular Biomedical Research Center, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Xiaohui Li
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - QiPing Feng
- Department of Medicine, Vanderbilt University, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Daniel I Chasman
- Harvard Medical School, Boston, MA, 02115, USA.,Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, 02215, USA
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA.,NHLBI Framingham Heart Study, Framingham, MA, 01702, USA
| | - Graham A Hitman
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ronald M Krauss
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr Way, Oakland, CA, 94609, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, WA, 98101, USA.,Kaiser Permanente Washington Health Research Institute, Seattle, WA, 98101, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA.,The Institute for Translational Genomics and Population Sciences, Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Saskia le Cessie
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.,Section of Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - C Michael Stein
- Department of Medicine, Vanderbilt University, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
323
|
Ahmad S, Mora S, Ridker PM, Hu FB, Chasman DI. Gene-Based Elevated Triglycerides and Type 2 Diabetes Mellitus Risk in the Women's Genome Health Study. Arterioscler Thromb Vasc Biol 2019; 39:97-106. [PMID: 30565958 DOI: 10.1161/atvbaha.118.311562] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective- Higher triglyceride (TG) is a risk factor for incident type 2 diabetes mellitus (T2DM), but paradoxically, genetic susceptibility for higher TG has been associated with lower T2DM risk. There is also evidence that the genetic association may be modified by baseline TG. Whether such associations can be replicated and the interaction is selective for certain TG-rich lipoprotein particles remains to be explored. Approach and Results- Cox regression involving TG, TG-rich lipoprotein particles, and genetic determinants of TG was performed among 15 813 participants with baseline fasting status in the WGHS (Women's Genome Health Study), including 1453 T2DM incident cases during a mean 18.6 (SD=5.3) years of follow-up. A weighted, 40-single-nucleotide polymorphism TG genetic risk score was inversely associated with incident T2DM (hazard ratio [95% CI], 0.66 [0.58-0.75]/10-TG risk alleles; P<0.0001) with adjustment for baseline body mass index, HDL (high-density lipoprotein) cholesterol, and TG. TG-associated risk was higher among individuals in the low compared with the high 40-single-nucleotide polymorphism TG genetic risk score tertile (hazard ratio [95% CI], 1.98 [1.83-2.14] versus 1.68 [1.58-1.80] per mmol/L; Pinteraction=0.0007). In TG-adjusted analysis, large and medium but not small TG-rich lipoprotein particles were associated with higher T2DM incidence for successively lower 40-single-nucleotide polymorphism TG genetic risk score tertiles, Pinteraction=0.013, 0.012, and 0.620 across tertiles, respectively. Conclusions- Our results confirm the previous observations of the paradoxical associations of TG with T2DM while focusing attention on the larger TG-rich lipoprotein particle subfractions, suggesting their importance in clinical profiling of T2DM risk.
Collapse
Affiliation(s)
- Shafqat Ahmad
- From the Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (S.A., F.B.H.).,Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.A., S.M., P.M.R., D.I.C.).,Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Sweden (S.A.)
| | - Samia Mora
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.A., S.M., P.M.R., D.I.C.).,Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.M., P.M.R.).,Center for Lipid Metabolomics, Harvard Medical School, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.M., P.M.R.)
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.A., S.M., P.M.R., D.I.C.).,Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.M., P.M.R.).,Center for Lipid Metabolomics, Harvard Medical School, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.M., P.M.R.)
| | - Frank B Hu
- From the Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (S.A., F.B.H.).,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (F.B.H.)
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.A., S.M., P.M.R., D.I.C.)
| |
Collapse
|
324
|
Gluchowski NL, Gabriel KR, Chitraju C, Bronson RT, Mejhert N, Boland S, Wang K, Lai ZW, Farese RV, Walther TC. Hepatocyte Deletion of Triglyceride-Synthesis Enzyme Acyl CoA: Diacylglycerol Acyltransferase 2 Reduces Steatosis Without Increasing Inflammation or Fibrosis in Mice. Hepatology 2019; 70:1972-1985. [PMID: 31081165 PMCID: PMC6893913 DOI: 10.1002/hep.30765] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/05/2019] [Indexed: 12/31/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by excess lipid accumulation in hepatocytes and represents a huge public health problem owing to its propensity to progress to nonalcoholic steatohepatitis, fibrosis, and liver failure. The lipids stored in hepatic steatosis (HS) are primarily triglycerides (TGs) synthesized by two acyl-CoA:diacylglycerol acyltransferase (DGAT) enzymes. Either DGAT1 or DGAT2 catalyzes this reaction, and these enzymes have been suggested to differentially utilize exogenous or endogenously synthesized fatty acids, respectively. DGAT2 has been linked to storage of fatty acids from de novo lipogenesis, a process increased in NAFLD. However, whether DGAT2 is more responsible for lipid accumulation in NAFLD and progression to fibrosis is currently unknown. Also, it is unresolved whether DGAT2 can be safely inhibited as a therapy for NAFLD. Here, we induced NAFLD-like disease in mice by feeding a diet rich in fructose, saturated fat, and cholesterol and found that hepatocyte-specific Dgat2 deficiency reduced expression of de novo lipogenesis genes and lowered liver TGs by ~70%. Importantly, the reduction in steatosis was not accompanied by increased inflammation or fibrosis, and insulin and glucose metabolism were unchanged. Conclusion: This study suggests that hepatic DGAT2 deficiency successfully reduces diet-induced HS and supports development of DGAT2 inhibitors as a therapeutic strategy for treating NAFLD and preventing downstream consequences.
Collapse
Affiliation(s)
- Nina L. Gluchowski
- Division of Gastroenterology and Nutrition, Boston Children’s Hospital, Boston, MA
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Katlyn R. Gabriel
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Boston, MA
| | - Chandramohan Chitraju
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | | | - Niklas Mejhert
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Sebastian Boland
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Kun Wang
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Zon Weng Lai
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Robert V. Farese
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| | - Tobias C. Walther
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Boston, MA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
325
|
Chen Z, Yu H, Shi X, Warren CR, Lotta LA, Friesen M, Meissner TB, Langenberg C, Wabitsch M, Wareham N, Benson MD, Gerszten RE, Cowan CA. Functional Screening of Candidate Causal Genes for Insulin Resistance in Human Preadipocytes and Adipocytes. Circ Res 2019; 126:330-346. [PMID: 31739742 DOI: 10.1161/circresaha.119.315246] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Rationale: Genome-wide association studies have identified genetic loci associated with insulin resistance (IR) but pinpointing the causal genes of a risk locus has been challenging. Objective: To identify candidate causal genes for IR, we screened regional and biologically plausible genes (16 in total) near the top 10 IR-loci in risk-relevant cell types, namely preadipocytes and adipocytes. Methods and Results: We generated 16 human Simpson-Golabi-Behmel syndrome preadipocyte knockout lines each with a single IR-gene knocked out by lentivirus-mediated CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 system. We evaluated each gene knockout by screening IR-relevant phenotypes in the 3 insulin-sensitizing mechanisms, including adipogenesis, lipid metabolism, and insulin signaling. We performed genetic analyses using data on the genotype-tissue expression portal expression quantitative trait loci database and accelerating medicines partnership type 2 diabetes mellitus Knowledge Portal to evaluate whether candidate genes prioritized by our in vitro studies were expression quantitative trait loci genes in human subcutaneous adipose tissue, and whether expression of these genes is associated with risk of IR, type 2 diabetes mellitus, and cardiovascular diseases. We further validated the functions of 3 new adipose IR genes by overexpression-based phenotypic rescue in the Simpson-Golabi-Behmel syndrome preadipocyte knockout lines. Twelve genes, PPARG, IRS-1, FST, PEPD, PDGFC, MAP3K1, GRB14, ARL15, ANKRD55, RSPO3, COBLL1, and LYPLAL1, showed diverse phenotypes in the 3 insulin-sensitizing mechanisms, and the first 7 of these genes could affect all the 3 mechanisms. Five out of 6 expression quantitative trait loci genes are among the top candidate causal genes and the abnormal expression levels of these genes (IRS-1, GRB14, FST, PEPD, and PDGFC) in human subcutaneous adipose tissue could be associated with increased risk of IR, type 2 diabetes mellitus, and cardiovascular disease. Phenotypic rescue by overexpression of the candidate causal genes (FST, PEPD, and PDGFC) in the Simpson-Golabi-Behmel syndrome preadipocyte knockout lines confirmed their function in adipose IR. Conclusions: Twelve genes showed diverse phenotypes indicating differential roles in insulin sensitization, suggesting mechanisms bridging the association of their genomic loci with IR. We prioritized PPARG, IRS-1, GRB14, MAP3K1, FST, PEPD, and PDGFC as top candidate genes. Our work points to novel roles for FST, PEPD, and PDGFC in adipose tissue, with consequences for cardiometabolic diseases.
Collapse
Affiliation(s)
- Zhifen Chen
- From the Beth Israel Deaconess Medical Center, Cardiovascular Institute, Harvard Medical School, Boston, MA (Z.C., H.Y., X.S., M.F., T.B.M., M.D.B., R.E.G, C.A.C.).,Harvard Stem Cell Institute, Harvard University, Cambridge, MA (Z.C., H.Y., M.F., C.R.W., T.B.M., C.A.C.)
| | - Haojie Yu
- From the Beth Israel Deaconess Medical Center, Cardiovascular Institute, Harvard Medical School, Boston, MA (Z.C., H.Y., X.S., M.F., T.B.M., M.D.B., R.E.G, C.A.C.).,Harvard Stem Cell Institute, Harvard University, Cambridge, MA (Z.C., H.Y., M.F., C.R.W., T.B.M., C.A.C.)
| | - Xu Shi
- From the Beth Israel Deaconess Medical Center, Cardiovascular Institute, Harvard Medical School, Boston, MA (Z.C., H.Y., X.S., M.F., T.B.M., M.D.B., R.E.G, C.A.C.)
| | - Curtis R Warren
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA (Z.C., H.Y., M.F., C.R.W., T.B.M., C.A.C.).,Cardiometabolic Disease Research, Boehringer-Ingelheim Pharmaceuticals, Inc, Ridgefield, CT (C.R.W.)
| | - Luca A Lotta
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom (L.A.L., C.L., N.W.)
| | - Max Friesen
- From the Beth Israel Deaconess Medical Center, Cardiovascular Institute, Harvard Medical School, Boston, MA (Z.C., H.Y., X.S., M.F., T.B.M., M.D.B., R.E.G, C.A.C.).,Harvard Stem Cell Institute, Harvard University, Cambridge, MA (Z.C., H.Y., M.F., C.R.W., T.B.M., C.A.C.)
| | - Torsten B Meissner
- From the Beth Israel Deaconess Medical Center, Cardiovascular Institute, Harvard Medical School, Boston, MA (Z.C., H.Y., X.S., M.F., T.B.M., M.D.B., R.E.G, C.A.C.).,Harvard Stem Cell Institute, Harvard University, Cambridge, MA (Z.C., H.Y., M.F., C.R.W., T.B.M., C.A.C.)
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom (L.A.L., C.L., N.W.)
| | - Martin Wabitsch
- Pediatrics and Adolescent Medicine, Ulm University Hospital, Germany (M.W.)
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom (L.A.L., C.L., N.W.)
| | - Mark D Benson
- From the Beth Israel Deaconess Medical Center, Cardiovascular Institute, Harvard Medical School, Boston, MA (Z.C., H.Y., X.S., M.F., T.B.M., M.D.B., R.E.G, C.A.C.)
| | - Rob E Gerszten
- From the Beth Israel Deaconess Medical Center, Cardiovascular Institute, Harvard Medical School, Boston, MA (Z.C., H.Y., X.S., M.F., T.B.M., M.D.B., R.E.G, C.A.C.)
| | - Chad A Cowan
- From the Beth Israel Deaconess Medical Center, Cardiovascular Institute, Harvard Medical School, Boston, MA (Z.C., H.Y., X.S., M.F., T.B.M., M.D.B., R.E.G, C.A.C.).,Harvard Stem Cell Institute, Harvard University, Cambridge, MA (Z.C., H.Y., M.F., C.R.W., T.B.M., C.A.C.)
| |
Collapse
|
326
|
Balliu B, Durrant M, Goede OD, Abell N, Li X, Liu B, Gloudemans MJ, Cook NL, Smith KS, Knowles DA, Pala M, Cucca F, Schlessinger D, Jaiswal S, Sabatti C, Lind L, Ingelsson E, Montgomery SB. Genetic regulation of gene expression and splicing during a 10-year period of human aging. Genome Biol 2019; 20:230. [PMID: 31684996 PMCID: PMC6827221 DOI: 10.1186/s13059-019-1840-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 09/27/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Molecular and cellular changes are intrinsic to aging and age-related diseases. Prior cross-sectional studies have investigated the combined effects of age and genetics on gene expression and alternative splicing; however, there has been no long-term, longitudinal characterization of these molecular changes, especially in older age. RESULTS We perform RNA sequencing in whole blood from the same individuals at ages 70 and 80 to quantify how gene expression, alternative splicing, and their genetic regulation are altered during this 10-year period of advanced aging at a population and individual level. We observe that individuals are more similar to their own expression profiles later in life than profiles of other individuals their own age. We identify 1291 and 294 genes differentially expressed and alternatively spliced with age, as well as 529 genes with outlying individual trajectories. Further, we observe a strong correlation of genetic effects on expression and splicing between the two ages, with a small subset of tested genes showing a reduction in genetic associations with expression and splicing in older age. CONCLUSIONS These findings demonstrate that, although the transcriptome and its genetic regulation is mostly stable late in life, a small subset of genes is dynamic and is characterized by a reduction in genetic regulation, most likely due to increasing environmental variance with age.
Collapse
Affiliation(s)
- Brunilda Balliu
- Department of Pathology, Stanford University School of Medicine, Stanford, USA.
| | - Matthew Durrant
- Department of Genetics, Stanford University School of Medicine, Stanford, USA
| | - Olivia de Goede
- Department of Genetics, Stanford University School of Medicine, Stanford, USA
| | - Nathan Abell
- Department of Genetics, Stanford University School of Medicine, Stanford, USA
| | - Xin Li
- Department of Pathology, Stanford University School of Medicine, Stanford, USA
| | - Boxiang Liu
- Department of Biology, Stanford University School of Medicine, Stanford, USA
| | | | - Naomi L Cook
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Kevin S Smith
- Department of Pathology, Stanford University School of Medicine, Stanford, USA
| | | | - Mauro Pala
- Dipartimento di Scienze Biomediche, Universita di Sassari, Sassari, Italy
| | - Francesco Cucca
- Dipartimento di Scienze Biomediche, Universita di Sassari, Sassari, Italy
| | | | - Siddhartha Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, USA
| | - Chiara Sabatti
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, USA
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA.
- Stanford Cardiovascular Institute, Stanford University, Stanford, USA.
- Stanford Diabetes Research Center, Stanford University, Stanford, USA.
| | - Stephen B Montgomery
- Department of Pathology, Stanford University School of Medicine, Stanford, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, USA.
| |
Collapse
|
327
|
Beijer K, Nowak C, Sundström J, Ärnlöv J, Fall T, Lind L. In search of causal pathways in diabetes: a study using proteomics and genotyping data from a cross-sectional study. Diabetologia 2019; 62:1998-2006. [PMID: 31446444 PMCID: PMC6805963 DOI: 10.1007/s00125-019-4960-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 06/06/2019] [Indexed: 12/12/2022]
Abstract
AIMS/HYPOTHESIS The pathogenesis of type 2 diabetes is not fully understood. We investigated whether circulating levels of preselected proteins were associated with the outcome 'diabetes' and whether these associations were causal. METHODS In 2467 individuals of the population-based, cross-sectional EpiHealth study (45-75 years, 50% women), 249 plasma proteins were analysed by the proximity extension assay technique. DNA was genotyped using the Illumina HumanCoreExome-12 v1.0 BeadChip. Diabetes was defined as taking glucose-lowering treatment or having a fasting plasma glucose of ≥7.0 mmol/l. The associations between proteins and diabetes were assessed using logistic regression. To investigate causal relationships between proteins and diabetes, a bidirectional two-sample Mendelian randomisation was performed based on large, genome-wide association studies belonging to the DIAGRAM and MAGIC consortia, and a genome-wide association study in the EpiHealth study. RESULTS Twenty-six proteins were positively associated with diabetes, including cathepsin D, retinal dehydrogenase 1, α-L-iduronidase, hydroxyacid oxidase 1 and galectin-4 (top five findings). Three proteins, lipoprotein lipase, IGF-binding protein 2 and paraoxonase 3 (PON-3), were inversely associated with diabetes. Fourteen of the proteins are novel discoveries. The Mendelian randomisation study did not disclose any significant causal effects between the proteins and diabetes in either direction that were consistent with the relationships found between the protein levels and diabetes. CONCLUSIONS/INTERPRETATION The 29 proteins associated with diabetes are involved in several physiological pathways, but given the power of the study no causal link was identified for those proteins tested in Mendelian randomisation. Therefore, the identified proteins are likely to be biomarkers for type 2 diabetes, rather than representing causal pathways.
Collapse
Affiliation(s)
- Kristina Beijer
- Department of Medical Sciences, Uppsala University, UCR, Dag Hammarskjölds väg 38, SE-751 83, Uppsala, Sweden.
| | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society, Division of Family Medicine and Primary Care, Karolinska Institute, Stockholm, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, UCR, Dag Hammarskjölds väg 38, SE-751 83, Uppsala, Sweden
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society, Division of Family Medicine and Primary Care, Karolinska Institute, Stockholm, Sweden
- School of Health and Social Sciences, Dalarna University, Falun, Sweden
| | - Tove Fall
- Department of Medical Sciences, Uppsala University, UCR, Dag Hammarskjölds väg 38, SE-751 83, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, UCR, Dag Hammarskjölds väg 38, SE-751 83, Uppsala, Sweden
| |
Collapse
|
328
|
Tari AR, Nauman J, Zisko N, Skjellegrind HK, Bosnes I, Bergh S, Stensvold D, Selbæk G, Wisløff U. Temporal changes in cardiorespiratory fitness and risk of dementia incidence and mortality: a population-based prospective cohort study. LANCET PUBLIC HEALTH 2019; 4:e565-e574. [DOI: 10.1016/s2468-2667(19)30183-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 12/30/2022]
|
329
|
Klarin D, Busenkell E, Judy R, Lynch J, Levin M, Haessler J, Aragam K, Chaffin M, Haas M, Lindström S, Assimes TL, Huang J, Min Lee K, Shao Q, Huffman JE, Kabrhel C, Huang Y, Sun YV, Vujkovic M, Saleheen D, Miller DR, Reaven P, DuVall S, Boden WE, Pyarajan S, Reiner AP, Trégouët DA, Henke P, Kooperberg C, Gaziano JM, Concato J, Rader DJ, Cho K, Chang KM, Wilson PWF, Smith NL, O'Donnell CJ, Tsao PS, Kathiresan S, Obi A, Damrauer SM, Natarajan P. Genome-wide association analysis of venous thromboembolism identifies new risk loci and genetic overlap with arterial vascular disease. Nat Genet 2019; 51:1574-1579. [PMID: 31676865 PMCID: PMC6858581 DOI: 10.1038/s41588-019-0519-3] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/24/2019] [Indexed: 12/22/2022]
Abstract
Venous thromboembolism is a significant cause of mortality1, yet its genetic determinants are incompletely defined. We performed a discovery genome-wide association study in the Million Veteran Program and UK Biobank, with testing of approximately 13 million DNA sequence variants for association with venous thromboembolism (26,066 cases and 624,053 controls) and meta-analyzed both studies, followed by independent replication with up to 17,672 venous thromboembolism cases and 167,295 controls. We identified 22 previously unknown loci, bringing the total number of venous thromboembolism-associated loci to 33, and subsequently fine-mapped these associations. We developed a genome-wide polygenic risk score for venous thromboembolism that identifies 5% of the population at an equivalent incident venous thromboembolism risk to carriers of the established factor V Leiden p.R506Q and prothrombin G20210A mutations. Our data provide mechanistic insights into the genetic epidemiology of venous thromboembolism and suggest a greater overlap among venous and arterial cardiovascular disease than previously thought.
Collapse
Affiliation(s)
- Derek Klarin
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Vascular Surgery and Endovascular Therapy, University of Florida School of Medicine, Gainesville, FL, USA
| | - Emma Busenkell
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Renae Judy
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julie Lynch
- Veterans Affairs Informatics and Computing Infrastructure, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, USA
- University of Massachusetts College of Nursing & Health Sciences, Boston, MA, USA
| | - Michael Levin
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffery Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Krishna Aragam
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mark Chaffin
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mary Haas
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sara Lindström
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Themistocles L Assimes
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jie Huang
- Massachusetts Veterans Epidemiology Research and Information Center, Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Kyung Min Lee
- Veterans Affairs Informatics and Computing Infrastructure, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, USA
- Center for Healthcare Organization and Implementation Research, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, USA
- Boston University School of Public Health, Department of Health Law, Policy & Management, Boston, MA, USA
| | - Qing Shao
- Center for Healthcare Organization and Implementation Research, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, USA
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center, Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Christopher Kabrhel
- Center for Vascular Emergencies, Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yunfeng Huang
- Department of Epidemiology, Emory University Rollins School of Public Health, Department of Biomedical Informatics Emory University School of Medicine, Atlanta, GA, USA
- Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
| | - 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
- Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
| | - Marijana Vujkovic
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Danish Saleheen
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Donald R Miller
- Center for Healthcare Organization and Implementation Research, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, USA
- Boston University School of Public Health, Department of Health Law, Policy & Management, Boston, MA, USA
| | - Peter Reaven
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, USA
| | - Scott DuVall
- Veterans Affairs Informatics and Computing Infrastructure, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, USA
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - William E Boden
- Massachusetts Veterans Epidemiology Research and Information Center, Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Saiju Pyarajan
- Massachusetts Veterans Epidemiology Research and Information Center, Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alex P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - David-Alexandre Trégouët
- Bordeaux Population Health Research Center (INSERM UMR S 1219), University of Bordeaux, Bordeaux, France
| | - Peter Henke
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center, Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - John Concato
- Clinical Epidemiology Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Daniel J Rader
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center, Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kyong-Mi Chang
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter W F Wilson
- Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
- Emory Clinical Cardiovascular Research Institute, Atlanta, GA, USA
| | - Nicholas L Smith
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Christopher J O'Donnell
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center, Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip S Tsao
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sekar Kathiresan
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Verve Therapeutics, Cambridge, MA, USA
| | - Andrea Obi
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Scott M Damrauer
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pradeep Natarajan
- Veterans Affairs Boston Healthcare System, Boston, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
330
|
Chen L, Chen XW, Huang X, Song BL, Wang Y, Wang Y. Regulation of glucose and lipid metabolism in health and disease. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1420-1458. [PMID: 31686320 DOI: 10.1007/s11427-019-1563-3] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/15/2019] [Indexed: 02/08/2023]
Abstract
Glucose and fatty acids are the major sources of energy for human body. Cholesterol, the most abundant sterol in mammals, is a key component of cell membranes although it does not generate ATP. The metabolisms of glucose, fatty acids and cholesterol are often intertwined and regulated. For example, glucose can be converted to fatty acids and cholesterol through de novo lipid biosynthesis pathways. Excessive lipids are secreted in lipoproteins or stored in lipid droplets. The metabolites of glucose and lipids are dynamically transported intercellularly and intracellularly, and then converted to other molecules in specific compartments. The disorders of glucose and lipid metabolism result in severe diseases including cardiovascular disease, diabetes and fatty liver. This review summarizes the major metabolic aspects of glucose and lipid, and their regulations in the context of physiology and diseases.
Collapse
Affiliation(s)
- Ligong Chen
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Yan Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Yiguo Wang
- MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
331
|
Kang M, Sung J. A genome-wide search for gene-by-obesity interaction loci of dyslipidemia in Koreans shows diverse genetic risk alleles. J Lipid Res 2019; 60:2090-2101. [PMID: 31662442 DOI: 10.1194/jlr.p119000226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/21/2019] [Indexed: 11/20/2022] Open
Abstract
Dyslipidemia is a well-established risk factor for CVD. Studies suggest that similar fat accumulation in a given population might result in different levels of dyslipidemia risk among individuals; for example, despite similar or leaner body composition compared with Caucasians, Asians of Korean descent experience a higher prevalence of dyslipidemia. These variations imply a possible role of gene-obesity interactions on lipid profiles. Genome-wide association studies have identified more than 500 loci regulating plasma lipids, but the interaction structure between genes and obesity traits remains unclear. We hypothesized that some loci modify the effects of obesity on dyslipidemia risk and analyzed extensive gene-environment interactions (G×Es) at genome-wide levels to search for replicated gene-obesity interactive SNPs. In four Korean cohorts (n = 18,025), we identified and replicated 20 gene-obesity interactions, including novel variants (SCN1A and SLC12A8) and known lipid-associated variants (APOA5, BUD13, ZNF259, and HMGCR). When we estimated the additional heritability of dyslipidemia by considering G×Es, the gain was substantial for triglycerides (TGs) but mild for LDL cholesterol (LDL-C) and total cholesterol (Total-C); the interaction explained up to 18.7% of TG, 2.4% of LDL-C, and 1.9% of Total-C heritability associated with waist-hip ratio. Our findings suggest that some individuals are prone to develop abnormal lipid profiles, particularly with regard to TGs, even with slight increases in obesity indices; ethnic diversities in the risk alleles might partly explain the differential dyslipidemia risk between populations. Research about these interacting variables may facilitate knowledge-based approaches to personalize health guidelines according to individual genetic profiles.
Collapse
Affiliation(s)
- Moonil Kang
- Division of Genome and Health Big Data, Department of Public Health Sciences Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Joohon Sung
- Division of Genome and Health Big Data, Department of Public Health Sciences Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea .,Institute of Health and Environment, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
332
|
Siewert KM, Voight BF. Bivariate Genome-Wide Association Scan Identifies 6 Novel Loci Associated With Lipid Levels and Coronary Artery Disease. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002239. [PMID: 30525989 DOI: 10.1161/circgen.118.002239] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Plasma lipid levels are heritable and genetically associated with risk of coronary artery disease (CAD). However, genome-wide association studies (GWAS) routinely analyze these traits independent of one another. Joint GWAS for two related phenotypes can lead to a higher powered analysis to detect variants contributing to both traits. METHODS We performed a bivariate GWAS to discover novel loci associated with both heart disease, using a CAD meta-analysis (122 733 cases and 424 528 controls), and lipid traits, using results from the Global Lipid Genetics Consortium (188 577 subjects). RESULTS We identified six previously unreported loci at genome-wide significance ( P<5×10-8), three which were associated with triglycerides and CAD, two which were associated with LDL (low-density lipoprotein) cholesterol and CAD, and one associated with total cholesterol and CAD. At several of our loci, the GWAS signals jointly localize with genetic variants associated with expression level changes for more than one neighboring genes, indicating that these loci may be affecting disease risk through regulatory activity. CONCLUSIONS We discovered six novel variants individually associated with both lipids and CAD.
Collapse
Affiliation(s)
- Katherine M Siewert
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia (K.M.S.)
| | - Benjamin F Voight
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (B.F.V.).,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (B.F.V.).,Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (B.F.V.)
| |
Collapse
|
333
|
Ference BA, Bhatt DL, Catapano AL, Packard CJ, Graham I, Kaptoge S, Ference TB, Guo Q, Laufs U, Ruff CT, Cupido A, Hovingh GK, Danesh J, Holmes MV, Smith GD, Ray KK, Nicholls SJ, Sabatine MS. Association of Genetic Variants Related to Combined Exposure to Lower Low-Density Lipoproteins and Lower Systolic Blood Pressure With Lifetime Risk of Cardiovascular Disease. JAMA 2019; 322:1381-1391. [PMID: 31475726 PMCID: PMC6724415 DOI: 10.1001/jama.2019.14120] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/16/2019] [Indexed: 01/21/2023]
Abstract
IMPORTANCE The relationship between exposure to lower low-density lipoprotein cholesterol (LDL-C) and lower systolic blood pressure (SBP) with the risk of cardiovascular disease has not been reliably quantified. OBJECTIVE To assess the association of lifetime exposure to the combination of both lower LDL-C and lower SBP with the lifetime risk of cardiovascular disease. DESIGN, SETTING, AND PARTICIPANTS Among 438 952 participants enrolled in the UK Biobank between 2006 and 2010 and followed up through 2018, genetic LDL-C and SBP scores were used as instruments to divide participants into groups with lifetime exposure to lower LDL-C, lower SBP, or both. Differences in plasma LDL-C, SBP, and cardiovascular event rates between the groups were compared to estimate associations with lifetime risk of cardiovascular disease. EXPOSURES Differences in plasma LDL-C and SBP compared with participants with both genetic scores below the median. Genetic risk scores higher than the median were associated with lower LDL-C and lower SBP. MAIN OUTCOMES AND MEASURES Odds ratio (OR) for major coronary events, defined as coronary death, nonfatal myocardial infarction, or coronary revascularization. RESULTS The mean age of the 438 952 participants was 65.2 years (range, 40.4-80.0 years), 54.1% were women, and 24 980 experienced a first major coronary event. Compared with the reference group, participants with LDL-C genetic scores higher than the median had 14.7-mg/dL lower LDL-C levels and an OR of 0.73 for major coronary events (95% CI, 0.70-0.75; P < .001). Participants with SBP genetic scores higher than the median had 2.9-mm Hg lower SBP and an OR of 0.82 for major coronary events (95% CI, 0.79-0.85, P < .001). Participants in the group with both genetic scores higher than the median had 13.9-mg/dL lower LDL-C, 3.1-mm Hg lower SBP, and an OR of 0.61 for major coronary events (95% CI, 0.59-0.64; P < .001). In a 4 × 4 factorial analysis, exposure to increasing genetic risk scores and lower LDL-C levels and SBP was associated with dose-dependent lower risks of major coronary events. In a meta-regression analysis, combined exposure to 38.67-mg/dL lower LDL-C and 10-mm Hg lower SBP was associated with an OR of 0.22 for major coronary events (95% CI, 0.17-0.26; P < .001), and 0.32 for cardiovascular death (95% CI, 0.25-0.40; P < .001). CONCLUSIONS AND RELEVANCE Lifelong genetic exposure to lower levels of low-density lipoprotein cholesterol and lower systolic blood pressure was associated with lower cardiovascular risk. However, these findings cannot be assumed to represent the magnitude of benefit achievable from treatment of these risk factors.
Collapse
Affiliation(s)
- Brian A. Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, United Kingdom
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Deepak L. Bhatt
- Brigham and Women’s Hospital Heart and Vascular Center, Harvard Medical School, Boston, Massachusetts
| | - Alberico L. Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Multimedica IRCCS, Milano, Italy
| | - Chris J. Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ian Graham
- School of Medicine, Trinity College, Dublin, Ireland
| | - Stephen Kaptoge
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Thatcher B. Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, United Kingdom
| | - Qi Guo
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, United Kingdom
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Ulrich Laufs
- Department of Cardiology, University of Leipzig, Leipzig, Germany
| | - Christian T. Ruff
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Arjen Cupido
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, United Kingdom
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - G. Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - John Danesh
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Michael V. Holmes
- MRC Population Health Research Unit, Clinical Trial Service Unit, and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Kausik K. Ray
- School of Public Health, Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College London, London, United Kingdom
| | | | - Marc S. Sabatine
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
334
|
Wang W, Liu W, Fidler T, Wang Y, Tang Y, Woods B, Welch C, Cai B, Silvestre-Roig C, Ai D, Yang YG, Hidalgo A, Soehnlein O, Tabas I, Levine RL, Tall AR, Wang N. Macrophage Inflammation, Erythrophagocytosis, and Accelerated Atherosclerosis in Jak2 V617F Mice. Circ Res 2019; 123:e35-e47. [PMID: 30571460 DOI: 10.1161/circresaha.118.313283] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
RATIONALE The mechanisms driving atherothrombotic risk in individuals with JAK2 V617F ( Jak2 VF) positive clonal hematopoiesis or myeloproliferative neoplasms are poorly understood. OBJECTIVE The goal of this study was to assess atherosclerosis and underlying mechanisms in hypercholesterolemic mice with hematopoietic Jak2 VF expression. METHODS AND RESULTS Irradiated low-density lipoprotein receptor knockout ( Ldlr-/-) mice were transplanted with bone marrow from wild-type or Jak2 VF mice and fed a high-fat high-cholesterol Western diet. Hematopoietic functions and atherosclerosis were characterized. After 7 weeks of Western diet, Jak2 VF mice showed increased atherosclerosis. Early atherosclerotic lesions showed increased neutrophil adhesion and content, correlating with lesion size. After 12 weeks of Western diet, Jak2 VF lesions showed increased complexity, with larger necrotic cores, defective efferocytosis, prominent iron deposition, and costaining of erythrocytes and macrophages, suggesting erythrophagocytosis. Jak2 VF erythrocytes were more susceptible to phagocytosis by wild-type macrophages and showed decreased surface expression of CD47, a "don't-eat-me" signal. Human JAK2VF erythrocytes were also more susceptible to erythrophagocytosis. Jak2 VF macrophages displayed increased expression and production of proinflammatory cytokines and chemokines, prominent inflammasome activation, increased p38 MAPK (mitogen-activated protein kinase) signaling, and reduced levels of MerTK (c-Mer tyrosine kinase), a key molecule mediating efferocytosis. Increased erythrophagocytosis also suppressed efferocytosis. CONCLUSIONS Hematopoietic Jak2 VF expression promotes early lesion formation and increased complexity in advanced atherosclerosis. In addition to increasing hematopoiesis and neutrophil infiltration in early lesions, Jak2 VF caused cellular defects in erythrocytes and macrophages, leading to increased erythrophagocytosis but defective efferocytosis. These changes promote accumulation of iron in plaques and increased necrotic core formation which, together with exacerbated proinflammatory responses, likely contribute to plaque instability.
Collapse
Affiliation(s)
- Wei Wang
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY
| | - Wenli Liu
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY.,Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Tianjin Medical University, China (W.L., D.A.)
| | - Trevor Fidler
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY
| | - Ying Wang
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY
| | - Yang Tang
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY
| | - Brittany Woods
- Human Oncology and Pathogenesis Program (B.W., R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY.,Leukemia Service, Department of Medicine (B.W., R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carrie Welch
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY
| | - Bishuang Cai
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY
| | - Carlos Silvestre-Roig
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (C.S.-R., A.H., O.S.)
| | - Ding Ai
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Tianjin Medical University, China (W.L., D.A.)
| | - Yong-Guang Yang
- Columbia Center for Translational Immunology (Y.-G.Y.), Columbia University Medical Center, New York, NY
| | - Andres Hidalgo
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (C.S.-R., A.H., O.S.).,Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (A.H.)
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (C.S.-R., A.H., O.S.).,Department of Physiology and Pharmacology (FyFa), Karolinska Institutet, Stockholm, Sweden (O.S.).,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S.)
| | - Ira Tabas
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY
| | - Ross L Levine
- Human Oncology and Pathogenesis Program (B.W., R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY.,Leukemia Service, Department of Medicine (B.W., R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alan R Tall
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY
| | - Nan Wang
- From the Division of Molecular Medicine, Department of Medicine (W.W., W.L., T.F., Y.W., Y.T., C.W., B.C., I.T., A.R.T., N.W.), Columbia University Medical Center, New York, NY
| |
Collapse
|
335
|
The transferability of lipid loci across African, Asian and European cohorts. Nat Commun 2019; 10:4330. [PMID: 31551420 PMCID: PMC6760173 DOI: 10.1038/s41467-019-12026-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/19/2019] [Indexed: 02/02/2023] Open
Abstract
Most genome-wide association studies are based on samples of European descent. We assess whether the genetic determinants of blood lipids, a major cardiovascular risk factor, are shared across populations. Genetic correlations for lipids between European-ancestry and Asian cohorts are not significantly different from 1. A genetic risk score based on LDL-cholesterol-associated loci has consistent effects on serum levels in samples from the UK, Uganda and Greece (r = 0.23-0.28, p < 1.9 × 10-14). Overall, there is evidence of reproducibility for ~75% of the major lipid loci from European discovery studies, except triglyceride loci in the Ugandan samples (10% of loci). Individual transferable loci are identified using trans-ethnic colocalization. Ten of fourteen loci not transferable to the Ugandan population have pleiotropic associations with BMI in Europeans; none of the transferable loci do. The non-transferable loci might affect lipids by modifying food intake in environments rich in certain nutrients, which suggests a potential role for gene-environment interactions.
Collapse
|
336
|
Tabassum R, Rämö JT, Ripatti P, Koskela JT, Kurki M, Karjalainen J, Palta P, Hassan S, Nunez-Fontarnau J, Kiiskinen TTJ, Söderlund S, Matikainen N, Gerl MJ, Surma MA, Klose C, Stitziel NO, Laivuori H, Havulinna AS, Service SK, Salomaa V, Pirinen M, Jauhiainen M, Daly MJ, Freimer NB, Palotie A, Taskinen MR, Simons K, Ripatti S. Genetic architecture of human plasma lipidome and its link to cardiovascular disease. Nat Commun 2019; 10:4329. [PMID: 31551469 PMCID: PMC6760179 DOI: 10.1038/s41467-019-11954-8] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/13/2019] [Indexed: 01/07/2023] Open
Abstract
Understanding genetic architecture of plasma lipidome could provide better insights into lipid metabolism and its link to cardiovascular diseases (CVDs). Here, we perform genome-wide association analyses of 141 lipid species (n = 2,181 individuals), followed by phenome-wide scans with 25 CVD related phenotypes (n = 511,700 individuals). We identify 35 lipid-species-associated loci (P <5 ×10-8), 10 of which associate with CVD risk including five new loci-COL5A1, GLTPD2, SPTLC3, MBOAT7 and GALNT16 (false discovery rate<0.05). We identify loci for lipid species that are shown to predict CVD e.g., SPTLC3 for CER(d18:1/24:1). We show that lipoprotein lipase (LPL) may more efficiently hydrolyze medium length triacylglycerides (TAGs) than others. Polyunsaturated lipids have highest heritability and genetic correlations, suggesting considerable genetic regulation at fatty acids levels. We find low genetic correlations between traditional lipids and lipid species. Our results show that lipidomic profiles capture information beyond traditional lipids and identify genetic variants modifying lipid levels and risk of CVD.
Collapse
Affiliation(s)
- Rubina Tabassum
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Joel T Rämö
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Pietari Ripatti
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jukka T Koskela
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mitja Kurki
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics and Genetic Analysis Platform, Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric & Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Juha Karjalainen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Priit Palta
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Shabbeer Hassan
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Javier Nunez-Fontarnau
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tuomo T J Kiiskinen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Sanni Söderlund
- Research Programs Unit, Diabetes & Obesity, University of Helsinki and Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Niina Matikainen
- Research Programs Unit, Diabetes & Obesity, University of Helsinki and Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
- Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | | | - Michal A Surma
- Lipotype GmbH, Dresden, Germany
- Łukasiewicz Research Network-PORT Polish Center for Technology Development, Stablowicka 147 Str., 54-066, Wroclaw, Poland
| | | | - Nathan O Stitziel
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aki S Havulinna
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Susan K Service
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Matti Pirinen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Helsinki Institute for Information Technology HIIT and Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
| | - Matti Jauhiainen
- National Institute for Health and Welfare, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Biomedicum, Helsinki, Finland
| | - Mark J Daly
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Nelson B Freimer
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Psychiatric & Neurodevelopmental Genetics Unit, Department of Psychiatry, Analytic and Translational Genetics Unit, Department of Medicine, and the Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Marja-Riitta Taskinen
- Research Programs Unit, Diabetes & Obesity, University of Helsinki and Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Kai Simons
- Lipotype GmbH, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland.
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
- Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
337
|
Khetarpal SA, Babb PL, Zhao W, Hancock-Cerutti WF, Brown CD, Rader DJ, Voight BF. Multiplexed Targeted Resequencing Identifies Coding and Regulatory Variation Underlying Phenotypic Extremes of High-Density Lipoprotein Cholesterol in Humans. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002070. [PMID: 29987113 DOI: 10.1161/circgen.117.002070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/09/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Genome-wide association studies have uncovered common variants at many loci influencing human complex traits, such as high-density lipoprotein cholesterol (HDL-C). However, the contribution of the identified genes is difficult to ascertain from current efforts interrogating common variants with small effects. Thus, there is a pressing need for scalable, cost-effective strategies for uncovering causal variants, many of which may be rare and noncoding. METHODS Here, we used a molecular inversion probe target capture approach to resequence both coding and regulatory regions at 7 HDL-C-associated loci in 797 individuals with extremely high HDL-C versus 735 low-to-normal HDL-C controls. Our targets included protein-coding regions of GALNT2, APOA5, APOC3, SCARB1, CCDC92, ZNF664, CETP, and LIPG (>9 kb) and proximate noncoding regulatory features (>42 kb). RESULTS Exome-wide genotyping in 1114 of the 1532 participants yielded a >90% genotyping concordance rate with molecular inversion probe-identified variants in ≈90% of participants. This approach rediscovered nearly all established genome-wide association studies associations in GALNT2, CETP, and LIPG loci with significant and concordant associations with HDL-C from our phenotypic extremes design at 0.1% of the sample size of lipid genome-wide association studies. In addition, we identified a novel, rare, CETP noncoding variant enriched in the extreme high HDL-C group (P<0.01, score test). CONCLUSIONS Our targeted resequencing of individuals at the HDL-C phenotypic extremes offers a novel, efficient, and cost-effective approach for identifying rare coding and noncoding variation differences in extreme phenotypes and supports the rationale for applying this methodology to uncover rare variation-particularly noncoding variation-underlying myriad complex traits.
Collapse
Affiliation(s)
- Sumeet A Khetarpal
- Department of Genetics (S.A.K., P.L.B., W.Z., W.F.H.-C., C.D.B., D.J.R.).,Department of Medicine (S.A.K., W.Z., W.F.H.-C., D.J.R.)
| | - Paul L Babb
- Department of Genetics (S.A.K., P.L.B., W.Z., W.F.H.-C., C.D.B., D.J.R.).,Department of Systems Pharmacology and Translational Therapeutics (P.L.B., B.F.V.)
| | - Wei Zhao
- Department of Genetics (S.A.K., P.L.B., W.Z., W.F.H.-C., C.D.B., D.J.R.).,Department of Medicine (S.A.K., W.Z., W.F.H.-C., D.J.R.).,Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Albert Einstein College of Medicine, Bronx, NY (W.Z.)
| | - William F Hancock-Cerutti
- Department of Genetics (S.A.K., P.L.B., W.Z., W.F.H.-C., C.D.B., D.J.R.).,Department of Medicine (S.A.K., W.Z., W.F.H.-C., D.J.R.)
| | - Christopher D Brown
- Department of Genetics (S.A.K., P.L.B., W.Z., W.F.H.-C., C.D.B., D.J.R.).,Department of Medicine (S.A.K., W.Z., W.F.H.-C., D.J.R.)
| | - Daniel J Rader
- Department of Genetics (S.A.K., P.L.B., W.Z., W.F.H.-C., C.D.B., D.J.R.) .,Institute for Translational Medicine and Therapeutics (D.J.R., B.F.V.)
| | - Benjamin F Voight
- Department of Systems Pharmacology and Translational Therapeutics (P.L.B., B.F.V.) .,Institute for Translational Medicine and Therapeutics (D.J.R., B.F.V.)
| |
Collapse
|
338
|
Natarajan P, Jaiswal S, Kathiresan S. Clonal Hematopoiesis: Somatic Mutations in Blood Cells and Atherosclerosis. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e001926. [PMID: 29987111 DOI: 10.1161/circgen.118.001926] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The most important prognostic factor for atherosclerotic cardiovascular disease is age, independent of all other recognized risk factors. Recently, exome sequence analyses showed that somatic mutations in blood cells, a process termed clonal hematopoiesis, are common and increase in prevalence with age, with at least 1 in 10 adults older than 70 years affected. Carriers of clonal hematopoiesis have been shown to be not only at heightened risk for hematologic malignancy but also at increased risk for atherosclerotic cardiovascular disease. Here, we review the prior literature of clonal selection and expansion of hematopoietic stem cells and the evidence supporting its causal association with atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Pradeep Natarajan
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N., S.K.). .,Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA (P.N., S.K.).,Department of Medicine, Harvard Medical School, Boston, MA (P.N., S.K.)
| | - Siddhartha Jaiswal
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N., S.K.).,Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA (P.N., S.K.).,Department of Medicine, Harvard Medical School, Boston, MA (P.N., S.K.)
| | | |
Collapse
|
339
|
Machado CM, Leite NC, França PH, Cardoso CR, Salles GF, Villela-Nogueira CA. PNPLA3 gene polymorphism in Brazilian patients with type 2 diabetes: A prognostic marker beyond liver disease? Nutr Metab Cardiovasc Dis 2019; 29:965-971. [PMID: 31377187 DOI: 10.1016/j.numecd.2019.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/06/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Genetic factors may impact nonalcoholic fatty liver disease (NAFLD) severity. We aimed to assess the prevalence of patatin-like phospholipase domain-containing 3 protein (PNPLA3) gene rs738409 C > G polymorphism in Brazilian individuals with type 2 diabetes and to investigate its association with liver disease severity, diabetic chronic degenerative complications, and metabolic control. METHODS AND RESULTS PNPLA3 genotyping was performed and classified as CC, CG, and GG. Clinical and laboratory data were obtained, including chronic degenerative diabetes complications. Liver stiffness and steatosis were evaluated by transient hepatic elastography with CAP using FibroScan®. Multiple logistic regression was performed to investigate the association of PNPLA3 G allele with clinical and laboratory variables and with hepatic fibrosis/steatosis. Three hundred three patients were included (118 male, mean age 59 ± 9.5 years). The G allele frequency was 32.5% (CC 47%, CG 41%, and GG 12%). Significant liver fibrosis and severe steatosis were diagnosed in 26% and 43% of patients, respectively. The variables independently associated with the G allele were coronary artery disease (OR: 2.25; 95% CI: 1.03-4.88; p = 0.04), better glycemic control (OR for having an HbA1c ≥ 8% [64 mmol/mol]: 0.53; 95% CI: 0.31-0.89; p = 0.01), and significant liver fibrosis (OR: 1.82; 95% CI: 1.04-3.17; p = 0.03). CONCLUSION In individuals with diabetes and NAFLD, PNPLA3 gene rs738409 C > G polymorphism is a marker for the risk of significant liver fibrosis and cardiovascular disease and may be associated with better glycemic control.
Collapse
Affiliation(s)
- Carolina M Machado
- School of Medicine, Internal Medicine Department and Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Nathalie C Leite
- School of Medicine, Internal Medicine Department and Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo H França
- University of Joinville, UNIVILLE, Joinville, Santa Catarina, Brazil
| | - Claudia R Cardoso
- School of Medicine, Internal Medicine Department and Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gil F Salles
- School of Medicine, Internal Medicine Department and Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cristiane A Villela-Nogueira
- School of Medicine, Internal Medicine Department and Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
340
|
Luukkonen PK, Nick A, Hölttä-Vuori M, Thiele C, Isokuortti E, Lallukka-Brück S, Zhou Y, Hakkarainen A, Lundbom N, Peltonen M, Orho-Melander M, Orešič M, Hyötyläinen T, Hodson L, Ikonen E, Yki-Järvinen H. Human PNPLA3-I148M variant increases hepatic retention of polyunsaturated fatty acids. JCI Insight 2019; 4:127902. [PMID: 31434800 PMCID: PMC6777808 DOI: 10.1172/jci.insight.127902] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
The common patatin-like phospholipase domain-containing protein 3 (PNPLA3) variant I148M predisposes to nonalcoholic liver disease but not its metabolic sequelae. We compared the handling of labeled polyunsaturated fatty acids (PUFAs) and saturated fatty acids (SFA) in vivo in humans and in cells harboring different PNPLA3 genotypes. In 148M homozygous individuals, triglycerides (TGs) in very low-density lipoproteins (VLDL) were depleted of PUFAs both under fasting and postprandial conditions compared with 148I homozygotes, and the PUFA/SFA ratio in VLDL-TGs was lower relative to the chylomicron precursor pool. In human PNPLA3-148M and PNPLA3-KO cells, PUFA but not SFA incorporation into TGs was increased at the expense of phosphatidylcholines, and under lipolytic conditions, PUFA-containing diacylglycerols (DAGs) accumulated compared with PNPLA3-148I cells. Polyunsaturated TGs were increased, while phosphatidylcholines (PCs) were decreased in the human liver in 148M homozygous individuals as compared with 148I homozygotes. We conclude that human PNPLA3-I148M is a loss-of-function allele that remodels liver TGs in a polyunsaturated direction by impairing hydrolysis/transacylation of PUFAs from DAGs to feed phosphatidylcholine synthesis.
Collapse
Affiliation(s)
- Panu K. Luukkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Auli Nick
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Faculty of Medicine, Department of Anatomy, University of Helsinki, Helsinki, Finland
| | - Maarit Hölttä-Vuori
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Faculty of Medicine, Department of Anatomy, University of Helsinki, Helsinki, Finland
| | | | - Elina Isokuortti
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Susanna Lallukka-Brück
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - You Zhou
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Antti Hakkarainen
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Department of Neuroscience and Biomedical Engineering, Aalto University School of Science, Espoo, Finland
| | - Nina Lundbom
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Markku Peltonen
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | | | - Matej Orešič
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- School of Medical Sciences and
| | | | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Elina Ikonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Faculty of Medicine, Department of Anatomy, University of Helsinki, Helsinki, Finland
| | - Hannele Yki-Järvinen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
341
|
Jaiswal S, Libby P. Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat Rev Cardiol 2019; 17:137-144. [DOI: 10.1038/s41569-019-0247-5] [Citation(s) in RCA: 249] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2019] [Indexed: 12/21/2022]
|
342
|
Xia MF, Bian H, Gao X. NAFLD and Diabetes: Two Sides of the Same Coin? Rationale for Gene-Based Personalized NAFLD Treatment. Front Pharmacol 2019; 10:877. [PMID: 31447675 PMCID: PMC6691129 DOI: 10.3389/fphar.2019.00877] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) has been increasing rapidly and at the forefront of worldwide concern. Characterized by excessive fat accumulation in the liver, NAFLD regularly coexists with metabolic disorders, including type 2 diabetes, obesity, and cardiovascular disease. It has been well established that the presence of NAFLD increases the incidence of type 2 diabetes, while diabetes aggravates NAFLD to more severe forms of steatohepatitis, cirrhosis, and hepatocellular carcinoma. However, recent progress on the genotype/phenotype relationships in NAFLD patients indicates the development of NAFLD with a relative conservation of glucose metabolism in individuals with specific gene variants, such as the patatin-like phospholipase domain-containing 3 (PNPLA3) and transmembrane 6 superfamily member 2 protein (TM6SF2) variants. This review will focus on the clinical and pathophysiological connections between NAFLD and type 2 diabetes and will also discuss a disproportionate progression of NAFLD and diabetes, and the different responses to lifestyle and drug intervention in NAFLD patients with specific gene variants that may give insight into personalized treatment for NAFLD.
Collapse
Affiliation(s)
- Ming-Feng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Hua Bian
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| |
Collapse
|
343
|
Locke AE, Steinberg KM, Chiang CWK, Service SK, Havulinna AS, Stell L, Pirinen M, Abel HJ, Chiang CC, Fulton RS, Jackson AU, Kang CJ, Kanchi KL, Koboldt DC, Larson DE, Nelson J, Nicholas TJ, Pietilä A, Ramensky V, Ray D, Scott LJ, Stringham HM, Vangipurapu J, Welch R, Yajnik P, Yin X, Eriksson JG, Ala-Korpela M, Järvelin MR, Männikkö M, Laivuori H, Dutcher SK, Stitziel NO, Wilson RK, Hall IM, Sabatti C, Palotie A, Salomaa V, Laakso M, Ripatti S, Boehnke M, Freimer NB. Exome sequencing of Finnish isolates enhances rare-variant association power. Nature 2019; 572:323-328. [PMID: 31367044 PMCID: PMC6697530 DOI: 10.1038/s41586-019-1457-z] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 07/02/2019] [Indexed: 12/30/2022]
Abstract
Exome-sequencing studies have generally been underpowered to identify deleterious alleles with a large effect on complex traits as such alleles are mostly rare. Because the population of northern and eastern Finland has expanded considerably and in isolation following a series of bottlenecks, individuals of these populations have numerous deleterious alleles at a relatively high frequency. Here, using exome sequencing of nearly 20,000 individuals from these regions, we investigate the role of rare coding variants in clinically relevant quantitative cardiometabolic traits. Exome-wide association studies for 64 quantitative traits identified 26 newly associated deleterious alleles. Of these 26 alleles, 19 are either unique to or more than 20 times more frequent in Finnish individuals than in other Europeans and show geographical clustering comparable to Mendelian disease mutations that are characteristic of the Finnish population. We estimate that sequencing studies of populations without this unique history would require hundreds of thousands to millions of participants to achieve comparable association power.
Collapse
Affiliation(s)
- Adam E Locke
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Karyn Meltz Steinberg
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Charleston W K Chiang
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Quantitative and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Susan K Service
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Aki S Havulinna
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Laurel Stell
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Matti Pirinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Helsinki Institute for Information Technology HIIT and Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
| | - Haley J Abel
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Colby C Chiang
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Robert S Fulton
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Chul Joo Kang
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Krishna L Kanchi
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel C Koboldt
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - David E Larson
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Joanne Nelson
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Thomas J Nicholas
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- USTAR Center for Genetic Discovery and Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Arto Pietilä
- National Institute for Health and Welfare, Helsinki, Finland
| | - Vasily Ramensky
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
- Federal State Institution "National Medical Research Center for Preventive Medicine" of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Debashree Ray
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Departments of Epidemiology and Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Pranav Yajnik
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Xianyong Yin
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Johan G Eriksson
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Ala-Korpela
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | - Marjo-Riitta Järvelin
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Minna Männikkö
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Northern Finland Birth Cohorts, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and University of Tampere, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Susan K Dutcher
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Nathan O Stitziel
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Richard K Wilson
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Ira M Hall
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Chiara Sabatti
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Analytical and Translational Genetics Unit (ATGU), Psychiatric & Neurodevelopmental Genetics Unit, Departments of Psychiatry and Neurology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| | - Nelson B Freimer
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
344
|
Abstract
Causal inference is essential across the biomedical, behavioural and social sciences.By progressing from confounded statistical associations to evidence of causal relationships, causal inference can reveal complex pathways underlying traits and diseases and help to prioritize targets for intervention. Recent progress in genetic epidemiology - including statistical innovation, massive genotyped data sets and novel computational tools for deep data mining - has fostered the intense development of methods exploiting genetic data and relatedness to strengthen causal inference in observational research. In this Review, we describe how such genetically informed methods differ in their rationale, applicability and inherent limitations and outline how they should be integrated in the future to offer a rich causal inference toolbox.
Collapse
|
345
|
Spracklen CN, Karaderi T, Yaghootkar H, Schurmann C, Fine RS, Kutalik Z, Preuss MH, Lu Y, Wittemans LBL, Adair LS, Allison M, Amin N, Auer PL, Bartz TM, Blüher M, Boehnke M, Borja JB, Bork-Jensen J, Broer L, Chasman DI, Chen YDI, Chirstofidou P, Demirkan A, van Duijn CM, Feitosa MF, Garcia ME, Graff M, Grallert H, Grarup N, Guo X, Haesser J, Hansen T, Harris TB, Highland HM, Hong J, Ikram MA, Ingelsson E, Jackson R, Jousilahti P, Kähönen M, Kizer JR, Kovacs P, Kriebel J, Laakso M, Lange LA, Lehtimäki T, Li J, Li-Gao R, Lind L, Luan J, Lyytikäinen LP, MacGregor S, Mackey DA, Mahajan A, Mangino M, Männistö S, McCarthy MI, McKnight B, Medina-Gomez C, Meigs JB, Molnos S, Mook-Kanamori D, Morris AP, de Mutsert R, Nalls MA, Nedeljkovic I, North KE, Pennell CE, Pradhan AD, Province MA, Raitakari OT, Raulerson CK, Reiner AP, Ridker PM, Ripatti S, Roberston N, Rotter JI, Salomaa V, Sandoval-Zárate AA, Sitlani CM, Spector TD, Strauch K, Stumvoll M, Taylor KD, Thuesen B, Tönjes A, Uitterlinden AG, Venturini C, Walker M, Wang CA, Wang S, Wareham NJ, Willems SM, Willems van Dijk K, Wilson JG, Wu Y, Yao J, Young KL, Langenberg C, Frayling TM, et alSpracklen CN, Karaderi T, Yaghootkar H, Schurmann C, Fine RS, Kutalik Z, Preuss MH, Lu Y, Wittemans LBL, Adair LS, Allison M, Amin N, Auer PL, Bartz TM, Blüher M, Boehnke M, Borja JB, Bork-Jensen J, Broer L, Chasman DI, Chen YDI, Chirstofidou P, Demirkan A, van Duijn CM, Feitosa MF, Garcia ME, Graff M, Grallert H, Grarup N, Guo X, Haesser J, Hansen T, Harris TB, Highland HM, Hong J, Ikram MA, Ingelsson E, Jackson R, Jousilahti P, Kähönen M, Kizer JR, Kovacs P, Kriebel J, Laakso M, Lange LA, Lehtimäki T, Li J, Li-Gao R, Lind L, Luan J, Lyytikäinen LP, MacGregor S, Mackey DA, Mahajan A, Mangino M, Männistö S, McCarthy MI, McKnight B, Medina-Gomez C, Meigs JB, Molnos S, Mook-Kanamori D, Morris AP, de Mutsert R, Nalls MA, Nedeljkovic I, North KE, Pennell CE, Pradhan AD, Province MA, Raitakari OT, Raulerson CK, Reiner AP, Ridker PM, Ripatti S, Roberston N, Rotter JI, Salomaa V, Sandoval-Zárate AA, Sitlani CM, Spector TD, Strauch K, Stumvoll M, Taylor KD, Thuesen B, Tönjes A, Uitterlinden AG, Venturini C, Walker M, Wang CA, Wang S, Wareham NJ, Willems SM, Willems van Dijk K, Wilson JG, Wu Y, Yao J, Young KL, Langenberg C, Frayling TM, Kilpeläinen TO, Lindgren CM, Loos RJF, Mohlke KL. Exome-Derived Adiponectin-Associated Variants Implicate Obesity and Lipid Biology. Am J Hum Genet 2019; 105:15-28. [PMID: 31178129 DOI: 10.1016/j.ajhg.2019.05.002] [Show More Authors] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/30/2019] [Indexed: 12/25/2022] Open
Abstract
Circulating levels of adiponectin, an adipocyte-secreted protein associated with cardiovascular and metabolic risk, are highly heritable. To gain insights into the biology that regulates adiponectin levels, we performed an exome array meta-analysis of 265,780 genetic variants in 67,739 individuals of European, Hispanic, African American, and East Asian ancestry. We identified 20 loci associated with adiponectin, including 11 that had been reported previously (p < 2 × 10-7). Comparison of exome array variants to regional linkage disequilibrium (LD) patterns and prior genome-wide association study (GWAS) results detected candidate variants (r2 > .60) spanning as much as 900 kb. To identify potential genes and mechanisms through which the previously unreported association signals act to affect adiponectin levels, we assessed cross-trait associations, expression quantitative trait loci in subcutaneous adipose, and biological pathways of nearby genes. Eight of the nine loci were also associated (p < 1 × 10-4) with at least one obesity or lipid trait. Candidate genes include PRKAR2A, PTH1R, and HDAC9, which have been suggested to play roles in adipocyte differentiation or bone marrow adipose tissue. Taken together, these findings provide further insights into the processes that influence circulating adiponectin levels.
Collapse
Affiliation(s)
- Cassandra N Spracklen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tugce Karaderi
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Department of Biological Sciences, Faculty of Arts and Sciences, Eastern Mediterranean University, Famagusta, Cyprus; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; DTU Health Technology, Technical University of Denmark, Lyngby 2800, Denmark
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter EX2 5DW, UK; Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rebecca S Fine
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Zoltan Kutalik
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter EX2 5DW, UK; University Center for Primary Care and Public Health, University of Lausanne, Lausanne 1010, Switzerland; Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN 37203-1738, USA; Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura B L Wittemans
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Linda S Adair
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Matthew Allison
- Department of Family Medicine and Public Health, University of California, San Diego, CA 92093, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015CN, the Netherlands
| | - Paul L Auer
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA; Department of Biostatistics, University of Washington, Seattle, WA 98101, USA
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 4103, Germany
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Judith B Borja
- Office of Population Studies Foundation, Inc, Cebu City, Philippines; Department of Nutrition and Dietetics, University of San Carlos, Cebu City, Philippines
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Linda Broer
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Paraskevi Chirstofidou
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Ayse Demirkan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015CN, the Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015CN, the Netherlands
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Melissa E Garcia
- National Heart, Lung, and Blood Institute, Bethesda, MD 20892, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Center for Genome Sciences, Chapel Hill, NC 27599, USA
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, München-Neuherberg 85764, Germany; German Center for Diabetes Research, München-Neuherberg 85765, Germany
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jeffrey Haesser
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jaeyoung Hong
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 2118, USA
| | - M Arfan Ikram
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Palo Alto, CA 94304, USA; Stanford Cardiovascular Institute, Stanford University of Medicine, Palo Alto, CA 94304, USA; Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala 75185, Sweden; Stanford Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
| | - Rebecca Jackson
- Division of Endocrinology, Diabetes, and Metabolism, Ohio State University, Columbus, OH 43210, USA
| | - Pekka Jousilahti
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere 33522, Finland; Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33522, Finland
| | - Jorge R Kizer
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 4103, Germany
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, München-Neuherberg 85764, Germany; German Center for Diabetes Research, München-Neuherberg 85765, Germany
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University of Hospital, Kuopio 70029 KYS, Finland
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado-Denver, Denver, CO 80045, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33522, Finland
| | - Jin Li
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala 75185, Sweden
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33522, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33521, Finland
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - David A Mackey
- Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA 6009, Australia; Centre for Ophthalmology and Visual Science, Lions Eye Institute, The University of Western Australia, Perth, WA 6009, Australia
| | - Anubha Mahajan
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK; NIHR Biomedical Research Centre, Guy's and St Thomas' Foundation Trust, London SE1 9RT, UK
| | - Satu Männistö
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Mark I McCarthy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK; Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7FZ, UK
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, WA 98101, USA
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands
| | - James B Meigs
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Program in Population and Medical Genetics, Broad Institute, Cambridge, MA 02114, USA
| | - Sophie Molnos
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, München-Neuherberg 85764, Germany; German Center for Diabetes Research, München-Neuherberg 85765, Germany
| | - Dennis Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands; Department of Public Health and Primary Care, Leiden University Medical Center, Leiden 2334 ZA, the Netherlands
| | - Andrew P Morris
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK
| | - Renee de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA; Data Tecnica International, Glen Echo, MD 20812, USA
| | - Ivana Nedeljkovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015CN, the Netherlands
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig E Pennell
- School of Medicine and Public Health, Faculty of Medicine and Health, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Aruna D Pradhan
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland; Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Chelsea K Raulerson
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alex P Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Samuli Ripatti
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Public Health, University of Helsinki, Helsinki 00014, Finland; Institute for Molecular Medicine Finland, Helsinki 00014, Finland
| | - Neil Roberston
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Veikko Salomaa
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki 00271, Finland
| | | | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany; Chair of Genetic Epidemiology, Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 4103, Germany
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Betina Thuesen
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen 2400, Denmark
| | - Anke Tönjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 4103, Germany
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands
| | - Cristina Venturini
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Mark Walker
- Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle, UK
| | - Carol A Wang
- School of Medicine and Public Health, Faculty of Medicine and Health, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Shuai Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 2118, USA
| | | | - Sara M Willems
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden 2333 ZA, the Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Ying Wu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter EX2 5DW, UK
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Cecilia M Lindgren
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LF, UK
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Ichan School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
346
|
Tada H, Takamura M, Kawashiri MA. Lipoprotein(a) as an Old and New Causal Risk Factor of Atherosclerotic Cardiovascular Disease. J Atheroscler Thromb 2019; 26:583-591. [PMID: 31061262 PMCID: PMC6629747 DOI: 10.5551/jat.rv17034] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 04/02/2019] [Indexed: 12/16/2022] Open
Abstract
Lipoprotein(a) [Lp(a)], discovered in 1963, has been associated with atherosclerotic cardiovascular disease (ASCVD) independent of other traditional risk factors, including LDL cholesterol. Lp(a) is an apolipoprotein B (apoB)-containing lipoprotein, which contains an LDL-like particle. Unlike LDL, which is a primary therapeutic target to decrease ASCVD, current guidelines recommend measuring Lp(a) for risk assessments because there is no clear evidence demonstrating the clinical benefit of decreasing Lp(a) using classical drugs such as niacin. However, recent Mendelian randomization studies indicate that Lp(a) causally correlates with ASCVD. In addition, novel drugs, including PCSK9 inhibitors, as well as antisense oligonucleotide for apo(a), have exhibited efficacy in decreasing Lp(a) substantially, invigorating a discussion whether Lp(a) could be a novel therapeutic target for further ASCVD risk reduction. This review aims to provide current understanding, and future perspectives, of Lp(a), which is currently considered a mere biomarker but may emerge as a novel therapeutic target in future clinical settings.
Collapse
Affiliation(s)
- Hayato Tada
- Department of Cardiology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Masayuki Takamura
- Department of Cardiology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Masa-aki Kawashiri
- Department of Cardiology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| |
Collapse
|
347
|
Aryal B, Price NL, Suarez Y, Fernández-Hernando C. ANGPTL4 in Metabolic and Cardiovascular Disease. Trends Mol Med 2019; 25:723-734. [PMID: 31235370 DOI: 10.1016/j.molmed.2019.05.010] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/13/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023]
Abstract
Alterations in circulating lipids and ectopic lipid deposition impact on the risk of developing cardiovascular and metabolic diseases. Lipoprotein lipase (LPL) hydrolyzes fatty acids (FAs) from triglyceride (TAG)-rich lipoproteins including very low density lipoproteins (VLDLs) and chylomicrons, and regulates their distribution to peripheral tissues. Angiopoietin-like 4 (ANGPTL4) mediates the inhibition of LPL activity under different circumstances. Accumulating evidence associates ANGPTL4 directly with the risk of atherosclerosis and type 2 diabetes (T2D). This review focuses on recent findings on the role of ANGPTL4 in metabolic and cardiovascular diseases. We highlight human and murine studies that explore ANGPTL4 functions in different tissues and how these effect disease development through possible autocrine and paracrine forms of regulation.
Collapse
Affiliation(s)
- Binod Aryal
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Integrative Cell Signaling and Neurobiology of Metabolism Program, Yale University School of Medicine, New Haven, CT, USA; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Nathan L Price
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Integrative Cell Signaling and Neurobiology of Metabolism Program, Yale University School of Medicine, New Haven, CT, USA; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Yajaira Suarez
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Integrative Cell Signaling and Neurobiology of Metabolism Program, Yale University School of Medicine, New Haven, CT, USA; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Integrative Cell Signaling and Neurobiology of Metabolism Program, Yale University School of Medicine, New Haven, CT, USA; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
348
|
Musunuru K, Bernstein D, Cole FS, Khokha MK, Lee FS, Lin S, McDonald TV, Moskowitz IP, Quertermous T, Sankaran VG, Schwartz DA, Silverman EK, Zhou X, Hasan AAK, Luo XZJ. Functional Assays to Screen and Dissect Genomic Hits: Doubling Down on the National Investment in Genomic Research. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002178. [PMID: 29654098 DOI: 10.1161/circgen.118.002178] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The National Institutes of Health have made substantial investments in genomic studies and technologies to identify DNA sequence variants associated with human disease phenotypes. The National Heart, Lung, and Blood Institute has been at the forefront of these commitments to ascertain genetic variation associated with heart, lung, blood, and sleep diseases and related clinical traits. Genome-wide association studies, exome- and genome-sequencing studies, and exome-genotyping studies of the National Heart, Lung, and Blood Institute-funded epidemiological and clinical case-control studies are identifying large numbers of genetic variants associated with heart, lung, blood, and sleep phenotypes. However, investigators face challenges in identification of genomic variants that are functionally disruptive among the myriad of computationally implicated variants. Studies to define mechanisms of genetic disruption encoded by computationally identified genomic variants require reproducible, adaptable, and inexpensive methods to screen candidate variant and gene function. High-throughput strategies will permit a tiered variant discovery and genetic mechanism approach that begins with rapid functional screening of a large number of computationally implicated variants and genes for discovery of those that merit mechanistic investigation. As such, improved variant-to-gene and gene-to-function screens-and adequate support for such studies-are critical to accelerating the translation of genomic findings. In this White Paper, we outline the variety of novel technologies, assays, and model systems that are making such screens faster, cheaper, and more accurate, referencing published work and ongoing work supported by the National Heart, Lung, and Blood Institute's R21/R33 Functional Assays to Screen Genomic Hits program. We discuss priorities that can accelerate the impressive but incomplete progress represented by big data genomic research.
Collapse
Affiliation(s)
- Kiran Musunuru
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.).
| | - Daniel Bernstein
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - F Sessions Cole
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Mustafa K Khokha
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Frank S Lee
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Shin Lin
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Thomas V McDonald
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Ivan P Moskowitz
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Thomas Quertermous
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Vijay G Sankaran
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - David A Schwartz
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Edwin K Silverman
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Xiaobo Zhou
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Ahmed A K Hasan
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| | - Xiao-Zhong James Luo
- Cardiovascular Institute, Department of Medicine (K.M.), Department of Genetics (K.M.), and Department of Pathology and Laboratory Medicine (F.S.L.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia. Department of Pediatrics (D.B.), Cardiovascular Institute (D.B., T.Q.), and Department of Medicine (T.Q.), Stanford University, CA. Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO (F.S.C.). St. Louis Children's Hospital, MO (F.S.C.). Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT (M.K.K.). Division of Cardiology, Department of Medicine, University of Washington, Seattle (S.L.). Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa, FL (T.V.M.). Department of Pediatrics (I.P.M.), Department of Pathology (I.P.M.), and Department of Human Genetics (I.P.M.), The University of Chicago, IL. Division of Hematology/ Oncology, Boston Children's Hospital, MA (V.G.S.). Department of Pediatric Oncology, Dana-Farber Cancer Institute (V.G.S.) and Channing Division of Network Medicine, Brigham and Women's Hospital (E.K.S., X.Z.), Harvard Medical School, Boston. Broad Institute of MIT and Harvard, Cambridge, MA (V.G.S.). University of Colorado, Aurora (D.A.S.). Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.A.K.H., X.-z.J.L.)
| |
Collapse
|
349
|
Harshfield EL, Koulman A, Ziemek D, Marney L, Fauman EB, Paul DS, Stacey D, Rasheed A, Lee JJ, Shah N, Jabeen S, Imran A, Abbas S, Hina Z, Qamar N, Mallick NH, Yaqoob Z, Saghir T, Rizvi SNH, Memon A, Rasheed SZ, Memon FUR, Qureshi IH, Ishaq M, Frossard P, Danesh J, Saleheen D, Butterworth AS, Wood AM, Griffin JL. An Unbiased Lipid Phenotyping Approach To Study the Genetic Determinants of Lipids and Their Association with Coronary Heart Disease Risk Factors. J Proteome Res 2019; 18:2397-2410. [PMID: 30887811 PMCID: PMC6558644 DOI: 10.1021/acs.jproteome.8b00786] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Direct infusion high-resolution mass spectrometry (DIHRMS) is a novel, high-throughput approach to rapidly and accurately profile hundreds of lipids in human serum without prior chromatography, facilitating in-depth lipid phenotyping for large epidemiological studies to reveal the detailed associations of individual lipids with coronary heart disease (CHD) risk factors. Intact lipid profiling by DIHRMS was performed on 5662 serum samples from healthy participants in the Pakistan Risk of Myocardial Infarction Study (PROMIS). We developed a novel semi-targeted peak-picking algorithm to detect mass-to-charge ratios in positive and negative ionization modes. We analyzed lipid partial correlations, assessed the association of lipid principal components with established CHD risk factors and genetic variants, and examined differences between lipids for a common genetic polymorphism. The DIHRMS method provided information on 360 lipids (including fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, and sterol lipids), with a median coefficient of variation of 11.6% (range: 5.4-51.9). The lipids were highly correlated and exhibited a range of associations with clinical chemistry biomarkers and lifestyle factors. This platform can provide many novel insights into the effects of physiology and lifestyle on lipid metabolism, genetic determinants of lipids, and the relationship between individual lipids and CHD risk factors.
Collapse
Affiliation(s)
- Eric L. Harshfield
- MRC/BHF
Cardiovascular Epidemiology Unit, Department of Public Health and
Primary Care, University of Cambridge, Cambridge CB1 8RN, U.K.,Stroke
Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, U.K.
| | - Albert Koulman
- Core
Metabolomics and Lipidomics Laboratory, National Institute for Health Research Cambridge Biomedical Research
Centre, Cambridge CB2 0QQ, U.K.
| | - Daniel Ziemek
- Inflammation
and Immunology, Pfizer Worldwide Research & Development, 10785 Berlin, Germany
| | - Luke Marney
- College
of Science and Engineering, Seattle University, Seattle, Washington 98122, United States
| | - Eric B. Fauman
- Genomic
Sciences and Technologies, Pfizer Worldwide Research & Development, Cambridge, Massachusetts 02139, United States
| | - Dirk S. Paul
- MRC/BHF
Cardiovascular Epidemiology Unit, Department of Public Health and
Primary Care, University of Cambridge, Cambridge CB1 8RN, U.K.
| | - David Stacey
- MRC/BHF
Cardiovascular Epidemiology Unit, Department of Public Health and
Primary Care, University of Cambridge, Cambridge CB1 8RN, U.K.
| | - Asif Rasheed
- Center
for Non-Communicable Diseases, Karachi 75300, Pakistan
| | - Jung-Jin Lee
- Department
of Medicine, Mayo Hospital, Lahore 54000, Pakistan
| | - Nabi Shah
- Center
for Non-Communicable Diseases, Karachi 75300, Pakistan,Department
of Pharmacy, COMSATS Institute of Information
Technology, Abbottabad 22060, Pakistan
| | - Sehrish Jabeen
- Center
for Non-Communicable Diseases, Karachi 75300, Pakistan
| | - Atif Imran
- Center
for Non-Communicable Diseases, Karachi 75300, Pakistan
| | - Shahid Abbas
- Department
of Cardiology, Faisalabad Institute of Cardiology, Faisalabad 38000, Pakistan
| | - Zoubia Hina
- Department
of Cardiology, Faisalabad Institute of Cardiology, Faisalabad 38000, Pakistan
| | - Nadeem Qamar
- National Institute of Cardiovascular Disorders, Karachi 75510, Pakistan
| | | | - Zia Yaqoob
- Karachi Institute of Heart Diseases, Karachi 75950, Pakistan
| | - Tahir Saghir
- National Institute of Cardiovascular Disorders, Karachi 75510, Pakistan
| | | | - Anis Memon
- National Institute of Cardiovascular Disorders, Karachi 75510, Pakistan
| | | | | | | | - Muhammad Ishaq
- Karachi Institute of Heart Diseases, Karachi 75950, Pakistan
| | | | - John Danesh
- MRC/BHF
Cardiovascular Epidemiology Unit, Department of Public Health and
Primary Care, University of Cambridge, Cambridge CB1 8RN, U.K.
| | - Danish Saleheen
- Center
for Non-Communicable Diseases, Karachi 75300, Pakistan,Department
of Biostatistics and Epidemiology, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Adam S. Butterworth
- MRC/BHF
Cardiovascular Epidemiology Unit, Department of Public Health and
Primary Care, University of Cambridge, Cambridge CB1 8RN, U.K.
| | - Angela M. Wood
- MRC/BHF
Cardiovascular Epidemiology Unit, Department of Public Health and
Primary Care, University of Cambridge, Cambridge CB1 8RN, U.K.
| | - Julian L. Griffin
- Department
of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, U.K.,E-mail: . Tel: +44 1223 764 922. Fax: +44 1223 333 345
| |
Collapse
|
350
|
de Vries PS, Brown MR, Bentley AR, Sung YJ, Winkler TW, Ntalla I, Schwander K, Kraja AT, Guo X, Franceschini N, Cheng CY, Sim X, Vojinovic D, Huffman JE, Musani SK, Li C, Feitosa MF, Richard MA, Noordam R, Aschard H, Bartz TM, Bielak LF, Deng X, Dorajoo R, Lohman KK, Manning AK, Rankinen T, Smith AV, Tajuddin SM, Evangelou E, Graff M, Alver M, Boissel M, Chai JF, Chen X, Divers J, Gandin I, Gao C, Goel A, Hagemeijer Y, Harris SE, Hartwig FP, He M, Horimoto ARVR, Hsu FC, Jackson AU, Kasturiratne A, Komulainen P, Kühnel B, Laguzzi F, Lee JH, Luan J, Lyytikäinen LP, Matoba N, Nolte IM, Pietzner M, Riaz M, Said MA, Scott RA, Sofer T, Stančáková A, Takeuchi F, Tayo BO, van der Most PJ, Varga TV, Wang Y, Ware EB, Wen W, Yanek LR, Zhang W, Zhao JH, Afaq S, Amin N, Amini M, Arking DE, Aung T, Ballantyne C, Boerwinkle E, Broeckel U, Campbell A, Canouil M, Charumathi S, Chen YDI, Connell JM, de Faire U, de las Fuentes L, de Mutsert R, de Silva HJ, Ding J, Dominiczak AF, Duan Q, Eaton CB, Eppinga RN, Faul JD, Fisher V, Forrester T, Franco OH, Friedlander Y, Ghanbari M, Giulianini F, et alde Vries PS, Brown MR, Bentley AR, Sung YJ, Winkler TW, Ntalla I, Schwander K, Kraja AT, Guo X, Franceschini N, Cheng CY, Sim X, Vojinovic D, Huffman JE, Musani SK, Li C, Feitosa MF, Richard MA, Noordam R, Aschard H, Bartz TM, Bielak LF, Deng X, Dorajoo R, Lohman KK, Manning AK, Rankinen T, Smith AV, Tajuddin SM, Evangelou E, Graff M, Alver M, Boissel M, Chai JF, Chen X, Divers J, Gandin I, Gao C, Goel A, Hagemeijer Y, Harris SE, Hartwig FP, He M, Horimoto ARVR, Hsu FC, Jackson AU, Kasturiratne A, Komulainen P, Kühnel B, Laguzzi F, Lee JH, Luan J, Lyytikäinen LP, Matoba N, Nolte IM, Pietzner M, Riaz M, Said MA, Scott RA, Sofer T, Stančáková A, Takeuchi F, Tayo BO, van der Most PJ, Varga TV, Wang Y, Ware EB, Wen W, Yanek LR, Zhang W, Zhao JH, Afaq S, Amin N, Amini M, Arking DE, Aung T, Ballantyne C, Boerwinkle E, Broeckel U, Campbell A, Canouil M, Charumathi S, Chen YDI, Connell JM, de Faire U, de las Fuentes L, de Mutsert R, de Silva HJ, Ding J, Dominiczak AF, Duan Q, Eaton CB, Eppinga RN, Faul JD, Fisher V, Forrester T, Franco OH, Friedlander Y, Ghanbari M, Giulianini F, Grabe HJ, Grove ML, Gu CC, Harris TB, Heikkinen S, Heng CK, Hirata M, Hixson JE, Howard BV, Ikram MA, InterAct Consortium, Jacobs DR, Johnson C, Jonas JB, Kammerer CM, Katsuya T, Khor CC, Kilpeläinen TO, Koh WP, Koistinen HA, Kolcic I, Kooperberg C, Krieger JE, Kritchevsky SB, Kubo M, Kuusisto J, Lakka TA, Langefeld CD, Langenberg C, Launer LJ, Lehne B, Lemaitre RN, Li Y, Liang J, Liu J, Liu K, Loh M, Louie T, Mägi R, Manichaikul AW, McKenzie CA, Meitinger T, Metspalu A, Milaneschi Y, Milani L, Mohlke KL, Mosley TH, Mukamal KJ, Nalls MA, Nauck M, Nelson CP, Sotoodehnia N, O'Connell JR, Palmer ND, Pazoki R, Pedersen NL, Peters A, Peyser PA, Polasek O, Poulter N, Raffel LJ, Raitakari OT, Reiner AP, Rice TK, Rich SS, Robino A, Robinson JG, Rose LM, Rudan I, Schmidt CO, Schreiner PJ, Scott WR, Sever P, Shi Y, Sidney S, Sims M, Smith BH, Smith JA, Snieder H, Starr JM, Strauch K, Tan N, Taylor KD, Teo YY, Tham YC, Uitterlinden AG, van Heemst D, Vuckovic D, Waldenberger M, Wang L, Wang Y, Wang Z, Wei WB, Williams C, Wilson G, Wojczynski MK, Yao J, Yu B, Yu C, Yuan JM, Zhao W, Zonderman AB, Becker DM, Boehnke M, Bowden DW, Chambers JC, Deary IJ, Esko T, Farrall M, Franks PW, Freedman BI, Froguel P, Gasparini P, Gieger C, Horta BL, Kamatani Y, Kato N, Kooner JS, Laakso M, Leander K, Lehtimäki T, Lifelines Cohort, Groningen, The Netherlands (Lifelines Cohort Study), Magnusson PKE, Penninx B, Pereira AC, Rauramaa R, Samani NJ, Scott J, Shu XO, van der Harst P, Wagenknecht LE, Wang YX, Wareham NJ, Watkins H, Weir DR, Wickremasinghe AR, Zheng W, Elliott P, North KE, Bouchard C, Evans MK, Gudnason V, Liu CT, Liu Y, Psaty BM, Ridker PM, van Dam RM, Kardia SLR, Zhu X, Rotimi CN, Mook-Kanamori DO, Fornage M, Kelly TN, Fox ER, Hayward C, van Duijn CM, Tai ES, Wong TY, Liu J, Rotter JI, Gauderman WJ, Province MA, Munroe PB, Rice K, Chasman DI, Cupples LA, Rao DC, Morrison AC. Multiancestry Genome-Wide Association Study of Lipid Levels Incorporating Gene-Alcohol Interactions. Am J Epidemiol 2019; 188:1033-1054. [PMID: 30698716 PMCID: PMC6545280 DOI: 10.1093/aje/kwz005] [Show More Authors] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 01/05/2019] [Accepted: 01/08/2019] [Indexed: 12/27/2022] Open
Abstract
A person's lipid profile is influenced by genetic variants and alcohol consumption, but the contribution of interactions between these exposures has not been studied. We therefore incorporated gene-alcohol interactions into a multiancestry genome-wide association study of levels of high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and triglycerides. We included 45 studies in stage 1 (genome-wide discovery) and 66 studies in stage 2 (focused follow-up), for a total of 394,584 individuals from 5 ancestry groups. Analyses covered the period July 2014-November 2017. Genetic main effects and interaction effects were jointly assessed by means of a 2-degrees-of-freedom (df) test, and a 1-df test was used to assess the interaction effects alone. Variants at 495 loci were at least suggestively associated (P < 1 × 10-6) with lipid levels in stage 1 and were evaluated in stage 2, followed by combined analyses of stage 1 and stage 2. In the combined analysis of stages 1 and 2, a total of 147 independent loci were associated with lipid levels at P < 5 × 10-8 using 2-df tests, of which 18 were novel. No genome-wide-significant associations were found testing the interaction effect alone. The novel loci included several genes (proprotein convertase subtilisin/kexin type 5 (PCSK5), vascular endothelial growth factor B (VEGFB), and apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 (APOBEC1) complementation factor (A1CF)) that have a putative role in lipid metabolism on the basis of existing evidence from cellular and experimental models.
Collapse
Affiliation(s)
- Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Yun J Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Ioanna Ntalla
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Xiuqing Guo
- Genomic Outcomes, Pediatrics, Institute for Translational Genomics and Population Sciences, LA BioMed at Harbor-UCLA Medical Center, Torrance, California
| | - Nora Franceschini
- Epidemiology, University of North Carolina Gilling School of Global Public Health, Chapel Hill, North Carolina
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Centre for Quantitative Medicine, Academic Medicine Research Institute, Ophthalmology & Visual Sciences Academic Clinical Program
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jennifer E Huffman
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Solomon K Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Changwei Li
- Epidemiology and Biostatistics, University of Georgia at Athens College of Public Health, Athens, Georgia
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Melissa A Richard
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Raymond Noordam
- Internal Medicine, Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Biostatistics and Medicine, University of Washington, Seattle, Washington
| | - Lawrence F Bielak
- Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Xuan Deng
- Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Kurt K Lohman
- Public Health Sciences, Biostatistical Sciences, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Alisa K Manning
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Salman M Tajuddin
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Mariaelisa Graff
- Epidemiology, University of North Carolina Gilling School of Global Public Health, Chapel Hill, North Carolina
| | - Maris Alver
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | | | - Jin Fang Chai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Xu Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, Sweden
| | - Jasmin Divers
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ilaria Gandin
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Yanick Hagemeijer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
- Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Fernando P Hartwig
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, RS, Brazil
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Fang-Chi Hsu
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan
| | | | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Federica Laguzzi
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joseph H Lee
- Sergievsky Center and Taub Institute, Columbia University Medical Center, New York, New York
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center – Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Nana Matoba
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maik Pietzner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK
| | - Muhammad Riaz
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - M Abdullah Said
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Tamar Sofer
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, Massachusetts
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Bamidele O Tayo
- Department of Public Health Sciences, Loyola University Chicago, Maywood, Illinois
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tibor V Varga
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Yajuan Wang
- Department of Population and Quantitative Health and Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Erin B Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lisa R Yanek
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Department of Cardiology, Ealing Hospital, Middlesex, United Kingdom
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Saima Afaq
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marzyeh Amini
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program
| | - Christie Ballantyne
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, Texas
- Houston Methodist Debakey Heart and Vascular Center, Houston, Texas
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, Texas
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas
| | - Ulrich Broeckel
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Archie Campbell
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Sabanayagam Charumathi
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Centre for Quantitative Medicine, Academic Medicine Research Institute, Ophthalmology & Visual Sciences Academic Clinical Program
| | - Yii-Der Ida Chen
- Genomic Outcomes, Pediatrics, Institute for Translational Genomics and Population Sciences, LA BioMed at Harbor-UCLA Medical Center, Torrance, California
| | - John M Connell
- Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - Ulf de Faire
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, Missouri
| | - Renée de Mutsert
- Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - H Janaka de Silva
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Jingzhong Ding
- Center on Diabetes, Obesity, and Metabolism, Gerontology and Geriatric Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Anna F Dominiczak
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Qing Duan
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Charles B Eaton
- Department of Family Medicine and Epidemiology, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Ruben N Eppinga
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Virginia Fisher
- Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Terrence Forrester
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, Jamaica
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Institute of Social and Preventive Medicine
| | - Yechiel Friedlander
- Braun School of Public Health, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hans J Grabe
- Department Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - C Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Sami Heikkinen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Khoo Teck Puat – National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Makoto Hirata
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - James E Hixson
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, Texas
| | - Barbara V Howard
- MedStar Health Research Institute, Hyattsville, Maryland
- Center for Clinical and Translational Sciences and Department of Medicine, Georgetown-Howard Universities, Washington, District of Columbia
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - David R Jacobs
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Craig Johnson
- Collaborative Health Studies Coordinating Center, University of Washington, Seattle, Washington
| | - Jost Bruno Jonas
- Department of Ophthalmology, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
- Beijing Institute of Ophthalmology, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Capital Medical University, Beijing, China
| | - Candace M Kammerer
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tomohiro Katsuya
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Chiea Chuen Khor
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Heikki A Koistinen
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine and Abdominal Center, Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | | | - Charles Kooperberg
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, Washington
| | - Jose E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute
| | - Steve B Kritchevsky
- Sticht Center for Health Aging and Alzheimer's Prevention, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Michiaki Kubo
- Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Timo A Lakka
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Carl D Langefeld
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Medicine, University of Washington, Seattle, Washington
| | - Yize Li
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Jingjing Liang
- Department of Population and Quantitative Health and Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kiang Liu
- Epidemiology, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Marie Loh
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Ani W Manichaikul
- Biostatistics Section, Center for Public Health Genomics, University of Virginia, School of Medicine, West Complex, Charlottesville, Virginia
| | - Colin A McKenzie
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, Jamaica
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | | | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Thomas H Mosley
- Geriatrics, Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kenneth J Mukamal
- General Medicine & Primary Care, Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Mike A Nalls
- Data Tecnica International, Glen Echo, Maryland
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, Washington
| | - Jeff R O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | | | - Raha Pazoki
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, Sweden
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- DZHK
| | - Patricia A Peyser
- Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Ozren Polasek
- Department of Public Health, Department of Medicine, University of Split, Split, Croatia
- Psychiatric Hospital “Sveti Ivan”, Zagreb, Croatia
- Gen-info Ltd, Zagreb, Croatia
| | - Neil Poulter
- School of Public Health, Imperial College London, London, United Kingdom
| | - Leslie J Raffel
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, California
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Alex P Reiner
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, Washington
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, School of Medicine, West Complex, Charlottesville, Virginia
| | - Antonietta Robino
- Institute for Maternal and Child Health – IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Jennifer G Robinson
- Department of Epidemiology and Medicine, University of Iowa, Iowa City, Iowa
| | - Lynda M Rose
- Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Carsten O Schmidt
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Pamela J Schreiner
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - William R Scott
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter Sever
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Yuan Shi
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Stephen Sidney
- Division of Research, Kaiser Permanente of Northern California, Oakland, California
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Jennifer A Smith
- Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
- Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU, Munich, Germany
| | - Nicholas Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kent D Taylor
- Genomic Outcomes, Pediatrics, Institute for Translational Genomics and Population Sciences, LA BioMed at Harbor-UCLA Medical Center, Torrance, California
| | - Yik Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Science and Engineering, National University of Singapore, Singapore, Singapore
- Department of Statistics and Applied Probability, National University of Singapore, Singapore, Singapore
| | - Yih Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Diana van Heemst
- Internal Medicine, Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Dragana Vuckovic
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Lihua Wang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Yujie Wang
- Epidemiology, University of North Carolina Gilling School of Global Public Health, Chapel Hill, North Carolina
| | - Zhe Wang
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Wen Bin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Christine Williams
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Gregory Wilson
- Jackson Heart Study, School of Public Health, Jackson State University, Jackson, Mississippi
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Jie Yao
- Genomic Outcomes, Pediatrics, Institute for Translational Genomics and Population Sciences, LA BioMed at Harbor-UCLA Medical Center, Torrance, California
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Caizheng Yu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Min Yuan
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wei Zhao
- Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Alan B Zonderman
- Behavioral Epidemiology Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Diane M Becker
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan
| | - Donald W Bowden
- Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Department of Cardiology, Ealing Hospital, Middlesex, United Kingdom
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Imperial College Healthcare NHS Trust, London, United Kingdom
- MRC-PHE Centre for Environment and Health, Imperial College London, London, United Kingdom
- NIHR Imperial College Biomedical Research Centre, Imperial College London, London, United Kingdom
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
- Psychology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Boston, Massachusetts
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Västerbotten, Sweden
- OCDEM, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Barry I Freedman
- Nephrology, Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Philippe Froguel
- CNRS UMR 8199, European Genomic Institute for Diabetes
- Department of Genomics of Common Disease, Imperial College London, London, United Kingdom
| | - Paolo Gasparini
- Department of Medical Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health – IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research
| | - Bernardo L Horta
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, Middlesex, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
- NIHR Imperial College Biomedical Research Centre, Imperial College London, London, United Kingdom
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Karin Leander
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center – Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, Sweden
| | - Brenda Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - James Scott
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ya Xing Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | | | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- MRC-PHE Centre for Environment and Health, Imperial College London, London, United Kingdom
- NIHR Imperial College Biomedical Research Centre, Imperial College London, London, United Kingdom
- Health Data Research UK
- UK Dementia Research Institute
| | - Kari E North
- Epidemiology, University of North Carolina Gilling School of Global Public Health, Chapel Hill, North Carolina
- Carolina Center of Genome Sciences, University of North Carolina, Chapel Hill, North Carolina
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ching-Ti Liu
- Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Yongmei Liu
- Public Health Sciences, Epidemiology and Prevention, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Epidemiology, Medicine and Health Services, University of Washington, Seattle, Washington
- Kaiser Permanente Washington, Health Research Institute, Seattle, Washington
| | - Paul M Ridker
- Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sharon L R Kardia
- Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health and Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Dennis O Mook-Kanamori
- Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
- Public Health and Primary Care, Leiden University Medical Center, Leiden, Leiden
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Tanika N Kelly
- Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Ervin R Fox
- Cardiology, Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program
| | - Jingmin Liu
- WHI CCC, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jerome I Rotter
- Genomic Outcomes, Pediatrics, Institute for Translational Genomics and Population Sciences, LA BioMed at Harbor-UCLA Medical Center, Torrance, California
| | - W James Gauderman
- Biostatistics, Preventive Medicine, University of Southern California, Los Angeles, California
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- NIHR Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, United Kingdom
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Daniel I Chasman
- Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - L Adrienne Cupples
- Biostatistics, Boston University School of Public Health, Boston, Massachusetts
- NHLBI Framingham Heart Study, Framingham, Massachusetts
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|