14 research outputs found

    Atrasentan and renal events in patients with type 2 diabetes and chronic kidney disease (SONAR): a double-blind, randomised, placebo-controlled trial

    Get PDF
    Background: Short-term treatment for people with type 2 diabetes using a low dose of the selective endothelin A receptor antagonist atrasentan reduces albuminuria without causing significant sodium retention. We report the long-term effects of treatment with atrasentan on major renal outcomes. Methods: We did this double-blind, randomised, placebo-controlled trial at 689 sites in 41 countries. We enrolled adults aged 18–85 years with type 2 diabetes, estimated glomerular filtration rate (eGFR)25–75 mL/min per 1·73 m 2 of body surface area, and a urine albumin-to-creatinine ratio (UACR)of 300–5000 mg/g who had received maximum labelled or tolerated renin–angiotensin system inhibition for at least 4 weeks. Participants were given atrasentan 0·75 mg orally daily during an enrichment period before random group assignment. Those with a UACR decrease of at least 30% with no substantial fluid retention during the enrichment period (responders)were included in the double-blind treatment period. Responders were randomly assigned to receive either atrasentan 0·75 mg orally daily or placebo. All patients and investigators were masked to treatment assignment. The primary endpoint was a composite of doubling of serum creatinine (sustained for ≥30 days)or end-stage kidney disease (eGFR <15 mL/min per 1·73 m 2 sustained for ≥90 days, chronic dialysis for ≥90 days, kidney transplantation, or death from kidney failure)in the intention-to-treat population of all responders. Safety was assessed in all patients who received at least one dose of their assigned study treatment. The study is registered with ClinicalTrials.gov, number NCT01858532. Findings: Between May 17, 2013, and July 13, 2017, 11 087 patients were screened; 5117 entered the enrichment period, and 4711 completed the enrichment period. Of these, 2648 patients were responders and were randomly assigned to the atrasentan group (n=1325)or placebo group (n=1323). Median follow-up was 2·2 years (IQR 1·4–2·9). 79 (6·0%)of 1325 patients in the atrasentan group and 105 (7·9%)of 1323 in the placebo group had a primary composite renal endpoint event (hazard ratio [HR]0·65 [95% CI 0·49–0·88]; p=0·0047). Fluid retention and anaemia adverse events, which have been previously attributed to endothelin receptor antagonists, were more frequent in the atrasentan group than in the placebo group. Hospital admission for heart failure occurred in 47 (3·5%)of 1325 patients in the atrasentan group and 34 (2·6%)of 1323 patients in the placebo group (HR 1·33 [95% CI 0·85–2·07]; p=0·208). 58 (4·4%)patients in the atrasentan group and 52 (3·9%)in the placebo group died (HR 1·09 [95% CI 0·75–1·59]; p=0·65). Interpretation: Atrasentan reduced the risk of renal events in patients with diabetes and chronic kidney disease who were selected to optimise efficacy and safety. These data support a potential role for selective endothelin receptor antagonists in protecting renal function in patients with type 2 diabetes at high risk of developing end-stage kidney disease. Funding: AbbVie

    Meta-analysis of genome-wide association studies identifies eight new loci for type 2 diabetes in east Asians

    Get PDF
    We conducted a three-stage genetic study to identify susceptibility loci for type 2 diabetes (T2D) in east Asian populations. We followed our stage 1 meta-analysis of eight T2D genome-wide association studies (6,952 cases with T2D and 11,865 controls) with a stage 2 in silico replication analysis (5,843 cases and 4,574 controls) and a stage 3 de novo replication analysis (12,284 cases and 13,172 controls). The combined analysis identified eight new T2D loci reaching genome-wide significance, which mapped in or near GLIS3, PEPD, FITM2-R3HDML-HNF4A, KCNK16, MAEA, GCC1-PAX4, PSMD6 and ZFAND3. GLIS3, which is involved in pancreatic beta cell development and insulin gene expression1,2, is known for its association with fasting glucose levels3,4. The evidence of an association with T2D for PEPD5 and HNF4A6,7 has been shown in previous studies. KCNK16 may regulate glucose-dependent insulin secretion in the pancreas. These findings, derived from an east Asian population, provide new perspectives on the etiology of T2D

    Genome-wide association study meta-analysis reveals transethnic replication of mean arterial and pulse pressure loci

    No full text
    We conducted a genome-wide association study meta-analysis of mean arterial pressure and pulse pressure among 26 600 East Asian participants (stage 1) followed by replication study of up to 28 783 participants (stage 2). For novel loci, statistical significance was determined by a P<5.0×10 in joint analysis of stage 1 and stage 2 data. For loci reported by the previous mean arterial and pulse pressure genome-wide association study meta-analysis in Europeans, evidence of transethnic replication was determined by consistency in effect direction and a Bonferroni-corrected P<1.4×10. No novel loci were identified by the current study. Five independent mean arterial pressure variants demonstrated robust evidence for transethnic replication including rs17249754 at ATP2B1 (P=7.5×10), rs2681492 at ATP2B1 (P=3.4×10), rs11191593 at NT5C2 (1.1×10), rs3824755 at CYP17A1 (P=1.2×10), and rs13149993 at FGF5 (P=2.4×10). Two additional variants showed suggestive evidence of transethnic replication (consistency in effect direction and P<0.05), including rs319690 at MAP4 (P=0.014) and rs1173771 at NPR3 (P=0.018). For pulse pressure, robust evidence of replication was identified for 2 independent variants, including rs17249754 at ATP2B1 (P=1.2×10) and rs11191593 at NT5C2 (P=1.1×10), with suggestive evidence of replication among an additional 2 variants including rs3824755 at CYP17A1 (P=6.1×10) and rs2681492 at ATP2B1 (P=9.0×10). Replicated variants demonstrated consistency in effect sizes between East Asian and European samples, with effect size differences ranging from 0.03 to 0.24 mm Hg for mean arterial pressure and from 0.03 to 0.21 mm Hg for pulse pressure. In conclusion, we present the first evidence of transethnic replication of several mean arterial and pulse pressure loci in an East Asian population

    Protein-Truncating Variants at the Cholesteryl Ester Transfer Protein Gene and Risk for Coronary Heart Disease

    No full text
    RATIONALE: Therapies that inhibit CETP (cholesteryl ester transfer protein) have failed to demonstrate a reduction in risk for coronary heart disease (CHD). Human DNA sequence variants that truncate the CETP gene may provide insight into the efficacy of CETP inhibition. OBJECTIVE: To test whether protein-truncating variants (PTVs) at the CETP gene were associated with plasma lipid levels and CHD. METHODS AND RESULTS: We sequenced the exons of the CETP gene in 58 469 participants from 12 case-control studies (18 817 CHD cases, 39 652 CHD-free controls). We defined PTV as those that lead to a premature stop, disrupt canonical splice sites, or lead to insertions/deletions that shift frame. We also genotyped 1 Japanese-specific PTV in 27561 participants from 3 case-control studies (14 286 CHD cases, 13 275 CHD-free controls). We tested association of CETP PTV carrier status with both plasma lipids and CHD. Among 58 469 participants with CETP gene-sequencing data available, average age was 51.5 years and 43% were women; 1 in 975 participants carried a PTV at the CETP gene. Compared with noncarriers, carriers of PTV at CETP had higher high-density lipoprotein cholesterol (effect size, 22.6 mg/dL; 95% confidence interval, 18-27; P<1.0×10-4), lower low-density lipoprotein cholesterol (-12.2 mg/dL; 95% confidence interval, -23 to -0.98; P=0.033), and lower triglycerides (-6.3%; 95% confidence interval, -12 to -0.22; P=0.043). CETP PTV carrier status was associated with reduced risk for CHD (summary odds ratio, 0.70; 95% confidence interval, 0.54-0.90; P=5.1×10-3). CONCLUSIONS: Compared with noncarriers, carriers of PTV at CETP displayed higher high-density lipoprotein cholesterol, lower low-density lipoprotein cholesterol, lower triglycerides, and lower risk for CHD

    Protein-Truncating Variants at the Cholesteryl Ester Transfer Protein Gene and Risk for Coronary Heart DiseaseNovelty and Significance

    No full text
    Rationale: Therapies that inhibit CETP (cholesteryl ester transfer protein) have failed to demonstrate a reduction in risk for coronary heart disease (CHD). Human DNA sequence variants that truncate the CETP gene may provide insight into the efficacy of CETP inhibition. Objective: To test whether protein-truncating variants (PTVs) at the CETP gene were associated with plasma lipid levels and CHD. Methods and Results: We sequenced the exons of the CETP gene in 58 469 participants from 12 case-control studies (18 817 CHD cases, 39 652 CHD-free controls). We defined PTV as those that lead to a premature stop, disrupt canonical splice sites, or lead to insertions/deletions that shift frame. We also genotyped 1 Japanese-specific PTV in 27561 participants from 3 case-control studies (14 286 CHD cases, 13 275 CHD-free controls). We tested association of CETP PTV carrier status with both plasma lipids and CHD. Among 58 469 participants with CETP gene-sequencing data available, average age was 51.5 years and 43% were women; 1 in 975 participants carried a PTV at the CETP gene. Compared with noncarriers, carriers of PTV at CETP had higher high-density lipoprotein cholesterol (effect size, 22.6 mg/dL; 95% confidence interval, 18-27; P<1.0×10-4), lower low-density lipoprotein cholesterol (-12.2 mg/dL; 95% confidence interval, -23 to -0.98; P=0.033), and lower triglycerides (-6.3%; 95% confidence interval, -12 to -0.22; P=0.043). CETP PTV carrier status was associated with reduced risk for CHD (summary odds ratio, 0.70; 95% confidence interval, 0.54-0.90; P=5.1×10-3). Conclusions: Compared with noncarriers, carriers of PTV at CETP displayed higher high-density lipoprotein cholesterol, lower low-density lipoprotein cholesterol, lower triglycerides, and lower risk for CHD

    Interethnic analyses of blood pressure loci in populations of East Asian and European descent

    No full text
    Blood pressure (BP) is a major risk factor for cardiovascular disease and more than 200 genetic loci associated with BP are known. Here, we perform a multi-stage genome-wide association study for BP (max N = 289,038) principally in East Asians and meta-analysis in East Asians and Europeans. We report 19 new genetic loci and ancestry-specific BP variants, conforming to a common ancestry-specific variant association model. At 10 unique loci, distinct non-rare ancestry-specific variants colocalize within the same linkage disequilibrium block despite the significantly discordant effects for the proxy shared variants between the ethnic groups. The genome-wide transethnic correlation of causal-variant effect-sizes is 0.898 and 0.851 for systolic and diastolic BP, respectively. Some of the ancestry-specific association signals are also influenced by a selective sweep. Our results provide new evidence for the role of common ancestry-specific variants and natural selection in ethnic differences in complex traits such as BP

    Meta-analysis of genome-wide association studies identifies eight new loci for type 2 diabetes in east Asians

    Get PDF
    We conducted a three-stage genetic study to identify susceptibility loci for type 2 diabetes (T2D) in east Asian populations. We followed our stage 1 meta-analysis of eight T2D genome-wide association studies (6,952 cases with T2D and 11,865 controls) with a stage 2 in silico replication analysis (5,843 cases and 4,574 controls) and a stage 3 de novo replication analysis (12,284 cases and 13,172 controls). The combined analysis identified eight new T2D loci reaching genome-wide significance, which mapped in or near GLIS3, PEPD, FITM2-R3HDML-HNF4A, KCNK16, MAEA, GCC1-PAX4, PSMD6 and ZFAND3. GLIS3, which is involved in pancreatic beta cell development and insulin gene expression, is known for its association with fasting glucose levels. The evidence of an association with T2D for PEPD and HNF4A has been shown in previous studies. KCNK16 may regulate glucose-dependent insulin secretion in the pancreas. These findings, derived from an east Asian population, provide new perspectives on the etiology of T2D

    Interethnic analyses of blood pressure loci in populations of East Asian and European descent

    No full text
    Abstract Blood pressure (BP) is a major risk factor for cardiovascular disease and more than 200 genetic loci associated with BP are known. Here, we perform a multi-stage genome-wide association study for BP (max N = 289,038) principally in East Asians and meta-analysis in East Asians and Europeans. We report 19 new genetic loci and ancestry-specific BP variants, conforming to a common ancestry-specific variant association model. At 10 unique loci, distinct non-rare ancestry-specific variants colocalize within the same linkage disequilibrium block despite the significantly discordant effects for the proxy shared variants between the ethnic groups. The genome-wide transethnic correlation of causal-variant effect-sizes is 0.898 and 0.851 for systolic and diastolic BP, respectively. Some of the ancestry-specific association signals are also influenced by a selective sweep. Our results provide new evidence for the role of common ancestry-specific variants and natural selection in ethnic differences in complex traits such as BP.Acknowledgements Marie Loh71,72 (Institute of Health Sciences, University of Oulu, P.O.Box 5000FI-90014 Oulu, Finland and Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK), Niek Verweij73 (Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands), Weihua Zhang72,74 (Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK and Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK), Benjamin Lehne72 (Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK), Irene Mateo Leach73 (Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands), Alexander Drong75 (Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK), James Abbott76 (Bioinformatics Support Service, Imperial College London, South Kensington, London SW7 2AZ, UK), Sian-Tsung Tan74,77 (Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK and National Heart and Lung Institute, Imperial College London, London W12 0NN, UK), William R. Scott72,77 (Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK and Lung Institute, Imperial College London, London W12 0NN, UK), Gianluca Campanella72 (Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK), Marc Chadeau-Hyam72 (Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK), Uzma Afzal72,74 (Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK and Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK), Tõnu Esko78,79,80,81 (Estonian Genome Center, University of Tartu, Riia 23c, 51010 Tartu, Estonia and Division of Endocrinology, Children’s Hospital Boston, Longwood 300, Boston, MA 02115, USA and Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142, USA), Sarah E. Harris82,83 (Medical Genetics Section, University of Edinburgh Molecular Medicine Centre and MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK and Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK), Jaana Hartiala84,85 (Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA 90033, USA and Institute for Genetic Medicine, USC Keck School of Medicine, Los Angeles, CA 90033, USA), Marcus E. Kleber86 (Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany), Richa Saxena87 (Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA), Alexandre F.R. Stewart88,89 (University of Ottawa Heart Institute, Cardiovascular Research Methods Centre, Ontario K1Y 4W7, Canada and Ruddy Canadian Cardiovascular Genetics Centre, Ontario K1Y 4W7, Canada), Tarunveer S. Ahluwalia90 (Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark), Imke Aits91 (Institute of Epidemiology and Biobank Popgen, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany), Alexessander Da Silva Couto Alves92 (Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPE) Centre for Environment and Health, School of Public Health, Imperial College London, London SW7 2AZ, UK), Shikta Das92 (Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPE) Centre for Environment and Health, School of Public Health, Imperial College London, London SW7 2AZ, UK), Jemma C. Hopewell93 (Clinical Trial Service Unit &amp; Epidemiological Studies Unit, University of Oxford, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK), Robert W. Koivula94 (Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, SE-205 02 Malmö, Sweden), Leo-Pekka Lyytikäinen95,96 (Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland and Department of Clinical Chemistry, University of Tampere School of Medicine, FI-33014 Tampere, Finland), Iris Postmus97,98 (Department of Gerontology and Geriatrics, Leiden University Medical Center, 2300 RC Leiden, Netherlands and Netherlands Consortium for Healthy Ageing, Leiden 2333 ZC, Netherlands), Olli T. Raitakari99,100 (Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, FI-20521 Turku, Finland and Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, FI-20520 Turku, Finland), Robert A. Scott101 (MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK), Rossella Sorice102 (Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, 80131 Naples, Italy), Vinicius Tragante103 (Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, Netherlands), Michela Traglia104,105 (Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milano, Italy and Institute for Maternal and Child Health—IRCCS ‘‘Burlo Garofolo’’—Trieste, 34137 Trieste, Italy), Jon White106 (UCL Genetics Institute, Department of Genetics, Environment and Evolution, UCL, London WC1E 6BT, UK), Inês Barroso107,108,109 (Metabolic Disease Group, The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK and University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK), Andrew Bjonnes87 (Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA), Rory Collins103 (Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, Netherlands), Gail Davies110 (Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh EH8 9JZ, UK), Graciela Delgado86 (Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany), Pieter A. Doevendans103 (Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, Netherlands), Lude Franke111 (Department of Genetics, University Medical Center, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands), Ron T. Gansevoort112 (Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands), Tanja B. Grammer86 (Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany), Niels Grarup86 (Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany), Jagvir Grewal72,74 (Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK and Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK), Anna-Liisa Hartikainen113,114 (Department of Obstetrics and Gynecology, University Hospital of Oulu, University of Oulu, Oulu FI-90014, Finland and Department of Clinical Sciences/Obsterics and Gynecology, University of Oulu, Oulu FI-90014, Finland), Stanley L. Hazen115,116 (Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, OH 44195, USA and Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA), Chris Hsu117 (Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA), Lise L.N. Husemoen118 (Research Centre for Prevention and Health, Glostrup University Hospital, 2600 Glostrup, Denmark), Johanne M. Justesen90 (Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark), Meena Kumari119 (Department of Epidemiology and Public Health, UCL, London WC1E 6BT, UK), Wolfgang Lieb91 (Institute of Epidemiology and Biobank Popgen, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany), David C.M. Liewald110 (Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh EH8 9JZ, UK), Evelin Mihailov78 (Estonian Genome Center, University of Tartu, Riia 23c, 51010 Tartu, Estonia), Lili Milani78 (Estonian Genome Center, University of Tartu, Riia 23c, 51010 Tartu, Estonia), Rebecca Mills74 (Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK), Nina Mononen95,96 (Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland and Department of Clinical Chemistry, University of Tampere School of Medicine, FI-33014 Tampere, Finland), Kjell Nikus120 (Heart Centre, Department of Cardiology, Tampere University Hospital, and University of Tampere School of Medicine, FI-33521 Tampere, Finland), Teresa Nutile102 (Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, 80131 Naples, Italy), Sarah Parish93 (Clinical Trial Service Unit &amp; Epidemiological Studies Unit, University of Oxford, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK), Olov Rolandsson121 (Department of Public Health &amp; Clinical Medicine, Section for Family Medicine, Umeå universitet, SE-901 85 Umeå, Sweden), Daniela Ruggiero102 (Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, 80131 Naples, Italy), Cinzia F. Sala104 (Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milano, Italy), Harold Snieder122 (Department of Epidemiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands), Thomas H.W. Spasø90 (Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark), Wilko Spiering123 (Department of Vascular Medicine, University Medical Center Utrecht, 3508 GA Utrecht, Netherlands), John M. Starr83,124 (Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK and Alzheimer Scotland Dementia Research Centre, University of Edinburgh, 7 George Square, Edinburgh EH8 9JZ, UK), David J. Stott125 (Academic Section of Geriatric Medicine, Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, Glasgow G4 0SF, UK), Daniel O. Stram117 (Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA), Silke Szymczak126 (Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel 24105, Germany), W.H.Wilson Tang115,116 (Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, OH 44195, USA and Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA), Stella Trompet127 (Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, Netherlands), Väinö Turjanmaa128,129 (Department of Clinical Physiology, Tampere University Hospital, FI-33521 Tampere, Finland and Department of Clinical Physiology, University of Tampere School of Medicine, FI-33014 Tampere, Finland), Marja Vaarasmaki130 (Department of Obstetrics and Gynecology, Oulu University Hospital, PO Box 23FI-90029 Oulu, Finland), Wiek H. van Gilst73 (Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands), Dirk J. van Veldhuisen73 (Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands), Jorma S. Viikari131,132 (Department of Medicine, Turku University Hospital, FI-20521 Turku, Finland and Department of Medicine, University of Turku, FI-20014 Turku, Finland), Folkert W. Asselbergs103,133,134 (Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, Netherlands and Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, 3511 GC Utrecht, Netherlands and Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London WC1E 6BT, UK), Marina Ciullo102 (Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, 80131 Naples, Italy), Andre Franke126 (Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel 24105, Germany), Paul W. Franks94,121,135 (Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, SE-205 02 Malmö, Sweden and Department of Public Health &amp; Clinical Medicine, Section for Family Medicine, Umeå universitet, SE-901 85 Umeå, Sweden and Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA), Steve Franks136 (Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London W120HS, UK), Myron D. Gross137 (School of Medicine, University of Minnesota, Minneapolis, MN 55455, USA), Torben Hansen90 (Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark), Marjo-Riitta Jarvelin72,92,138,139,140 (Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK and Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPE) Centre for Environment and Health, School of Public Health, Imperial College London, London SW7 2AZ, UK and Biocenter Oulu, University of Oulu, P.O. Box 5000 Aapistie 5A, FI-90014 Oulu, Finland and Unit of Primary Care, Oulu University Hospital, Kajaanintie 50 P.O.Box 20FI-90220 Oulu, Finland and Department of Children and Young People and Families, National Institute for Health and Welfare, Aapistie 1, Box 310, FI-90101 Oulu, Finland), Torben Jørgensen118 (Research Centre for Prevention and Health, Glostrup University Hospital, 2600 Glostrup, Denmark), Wouter J. Jukema127,133,141 (Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, Netherlands and Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, 3511 GC Utrecht, Netherlands and Interuniversity Cardiology Institute of the Netherlands, Utrecht 3511 EP, Netherlands), Mika Kähönen128,129 (Department of Clinical Physiology, Tampere University Hospital, FI-33521 Tampere, Finland and Department of Clinical Physiology, University of Tampere School of Medicine, FI-33014 Tampere, Finland), Mika Kivimaki119 (Department of Epidemiology and Public Health, UCL, London WC1E 6BT, UK), Terho Lehtimäki95,96 (Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland and Department of Clinical Chemistry, University of Tampere School of Medicine, FI-33014 Tampere, Finland), Allan Linneberg118 (Research Centre for Prevention and Health, Glostrup University Hospital, 2600 Glostrup, Denmark), Oluf Pedersen90 (Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark), Nilesh J. Samani142,143 (Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK), Daniela Toniolo104,144 (Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milano, Italy and Institute of Molecular GeneticsCNR, 27100 Pavia, Italy), Hooman Allayee84,85 (Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA 90033, USA and Institute for Genetic Medicine, USC Keck School of Medicine, Los Angeles, CA 90033, USA), Ian J. Deary83,110 (Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK and Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh EH8 9JZ, UK), Winfried März86,145,146 (Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany and Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria and Synlab Academy, Synlab Services GmbH, Gottlieb-Daimler-Straße 25, 68165 Mannheim, Germany), Andres Metspalu78 (Estonian Genome Center, University of Tartu, Riia 23c, 51010 Tartu, Estonia), Cisca Wijmenga111 (Department of Genetics, University Medical Center, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands), Bruce H.W. Wolffenbuttel147 (Department of Endocrinology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands), Paolo Vineis72 (Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK), Soterios A. KyrtopoulosNational Hellenic Research Foundation, Institute of Biological Research and Biotechnology, Athens 116 35, Greece), Jos C.S. Kleinjans149 (Department of Toxicogenomics, Maastricht University, Universiteitssingel 50, 6229ER Maastricht, Netherlands), Mark I. McCarthy75,150 (Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK and Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK), James Scott77 (National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
    corecore