2001
|
Allen NB, Lloyd-Jones D, Hwang SJ, Rasmussen-Torvik L, Fornage M, Morrison AC, Baldridge AS, Boerwinkle E, Levy D, Cupples LA, Fox CS, Thanassoulis G, Dufresne L, Daviglus M, Johnson AD, Reis J, Rotter J, Palmas W, Allison M, Pankow JS, O'Donnell CJ. Genetic loci associated with ideal cardiovascular health: A meta-analysis of genome-wide association studies. Am Heart J 2016; 175:112-20. [PMID: 27179730 PMCID: PMC4873714 DOI: 10.1016/j.ahj.2015.12.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 12/31/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Multiple genetic loci are associated with clinical cardiovascular (CV) disease and individual CV risk factors. Individuals with ideal levels of all major CV risk factors have very low risk for CV disease morbidity or mortality. Ideal levels of risk factors can be attained by lifestyle modifications; however, little is known about gene variants associated with ideal CV health. Our objective was to carry out a genome-wide association study on the trait. METHODS AND RESULTS We examined 2 dichotomous phenotypes of ideal CV health-clinical (untreated cholesterol <200 mg/dL, untreated blood pressure <120/<80, not diabetic) and clinical+behavioral (clinical plus: not a current smoker, body mass index <25 kg/m(2))-among white participants aged 50±5 years. We performed a meta-analysis of 4 genome-wide association studies (total n=11,708) from the MESA, CARDIA, ARIC, and Framingham Heart Study cohorts. We identified a single-nucleotide polymorphism (rs445925) in the APOC1/APOE region that was associated with clinical ideal CV health at genome-wide level of significance (P<2.0 × 10(-9)). The significance of this region was validated using exome chip genotyping. The association with ideal CV health was attenuated after adjusting for low-density lipoprotein cholesterol. CONCLUSION A common single-nucleotide polymorphism in the APOC1/APOE region, previously found to be associated with protective levels of cholesterol and lower CV risk, may be associated with ideal health. In future replication studies, larger sample sizes may be needed to detect loci with more modest effects on ideal CV health. In addition to the important impact of lifestyle modifications, we have identified evidence for gene variation that plays a role in ideal CV health.
Collapse
Affiliation(s)
- Norrina B Allen
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL.
| | - Donald Lloyd-Jones
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Shih-Jen Hwang
- Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), Framingham, MA; NHLBI's Framingham Heart Study, Framingham, MA
| | - Laura Rasmussen-Torvik
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Myriam Fornage
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX
| | - Alanna C Morrison
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX
| | - Abigail S Baldridge
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX
| | - Daniel Levy
- Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), Framingham, MA; NHLBI's Framingham Heart Study, Framingham, MA
| | | | - Caroline S Fox
- Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), Framingham, MA; NHLBI's Framingham Heart Study, Framingham, MA
| | - George Thanassoulis
- Department of Medicine and the Research Institute, Preventive and Genomic Cardiology, McGill University Health Center, Montreal, QC, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Line Dufresne
- Department of Medicine and the Research Institute, Preventive and Genomic Cardiology, McGill University Health Center, Montreal, QC, Canada
| | | | - Andrew D Johnson
- Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), Framingham, MA; NHLBI's Framingham Heart Study, Framingham, MA
| | - Jared Reis
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD
| | - Jerome Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Walter Palmas
- Department of Medicine, Columbia University, New York, NY
| | - Mathew Allison
- Division of Preventive Medicine, University of California, San Diego, CA
| | | | - Christopher J O'Donnell
- Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), Framingham, MA; NHLBI's Framingham Heart Study, Framingham, MA; Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
2002
|
Zhang H, Mo XB, Xu T, Lei SF, Zhang YH. Detecting novel genes for low-density lipoprotein cholesterol in European population using bioinformatics analysis. Per Med 2016; 13:225-231. [PMID: 29767610 DOI: 10.2217/pme.16.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM The aim of this study was to identify related genes for low-density lipoprotein cholesterol and evaluate the functional relevance to provide evidences for prioritizing these genes. MATERIALS & METHODS We performed a gene-based association study in about 188,578 individuals. Furthermore, we performed bioinformatics analyses to support the identified genes. RESULTS A total of 292 genes were found to be significant after Bonferroni correction (p < 2.3 × 10-6). Among these genes, 59 seemed to be associated with coronary artery disease (CAD). CONCLUSION The evidence obtained from the analyses of this study signified the importance of many genes, for example, LDLR, ABCG5, ABCG8, APOB, HNF1A, PTPN11, APOA5 and MCM6, which were also associated with CAD. The findings might provide more insights into the genetic basis of lipid metabolism and CAD.
Collapse
Affiliation(s)
- Huan Zhang
- Jiangsu Key Laboratory of Preventive & Translational Medicine for Geriatric Diseases, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, PR China.,Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu, Jiangsu 215123, PR China
| | - Xing-Bo Mo
- Jiangsu Key Laboratory of Preventive & Translational Medicine for Geriatric Diseases, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, PR China.,Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu, Jiangsu 215123, PR China.,Center for Genetic Epidemiology & Genomics, School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu, Jiangsu 215123, PR China
| | - Tan Xu
- Jiangsu Key Laboratory of Preventive & Translational Medicine for Geriatric Diseases, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, PR China.,Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu, Jiangsu 215123, PR China
| | - Shu-Feng Lei
- Jiangsu Key Laboratory of Preventive & Translational Medicine for Geriatric Diseases, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, PR China.,Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu, Jiangsu 215123, PR China.,Center for Genetic Epidemiology & Genomics, School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu, Jiangsu 215123, PR China
| | - Yong-Hong Zhang
- Jiangsu Key Laboratory of Preventive & Translational Medicine for Geriatric Diseases, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, PR China.,Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu, Jiangsu 215123, PR China
| |
Collapse
|
2003
|
Abstract
Although disproportionately affected by increasing rates of type 2 diabetes and dyslipidemias, Hispanic populations are underrepresented in efforts to understand genetic susceptibility to these disorders. Where research has been undertaken, these populations have provided substantial insight into identification of novel risk-associated genes and have aided in the ability to fine map previously described risk loci. Genome-wide analyses in Hispanic and trans-ethnic populations have resulted in identification of more than 40 replicated or novel genes with significant effects for type 2 diabetes or lipid traits. Initial investigations into rare variant effects have identified new risk-associated variants private to Hispanic populations, and preliminary results suggest metagenomic approaches in Hispanic populations, such as characterizing the gut microbiome, will enable the development of new predictive tools and therapeutic targets for type 2 diabetes. Future genome-wide studies in expanded cohorts of Hispanics are likely to result in new insights into the genetic etiology of metabolic health.
Collapse
Affiliation(s)
- Jennifer E Below
- The Human Genetics Center, University of Texas School of Public Health, Houston, TX, USA.
| | - Esteban J Parra
- Department of Anthropology, University of Toronto at Mississauga, Mississauga, ON, Canada
| |
Collapse
|
2004
|
Profiling and Validation of the Circular RNA Repertoire in Adult Murine Hearts. GENOMICS PROTEOMICS & BIOINFORMATICS 2016; 14:216-23. [PMID: 27132142 PMCID: PMC4996846 DOI: 10.1016/j.gpb.2016.02.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/19/2016] [Accepted: 02/15/2016] [Indexed: 11/22/2022]
Abstract
For several decades, cardiovascular disease has been the leading cause of death throughout all countries. There is a strong genetic component to many disease subtypes (e.g., cardiomyopathy) and we are just beginning to understand the relevant genetic factors. Several studies have related RNA splicing to cardiovascular disease and circular RNAs (circRNAs) are an emerging player. circRNAs, which originate through back-splicing events from primary transcripts, are resistant to exonucleases and typically not polyadenylated. Initial functional studies show clear phenotypic outcomes for selected circRNAs. We provide, for the first time, a comprehensive catalogue of RNase R-resistant circRNA species for the adult murine heart. This work combines state-of-the-art circle sequencing with our novel DCC software to explore the circRNA landscape of heart tissue. Overall, we identified 575 circRNA species that pass a beta-binomial test for enrichment (false discovery rate of 1%) in the exonuclease-treated sequencing sample. Several circRNAs can be directly attributed to host genes that have been previously described as associated with cardiovascular disease. Further studies of these candidate circRNAs may reveal disease-relevant properties or functions of specific circRNAs.
Collapse
|
2005
|
Zhao H, Fan D, Nyholt DR, Yang Y. Enrichment of SNPs in Functional Categories Reveals Genes Affecting Complex Traits. Hum Mutat 2016; 37:820-6. [PMID: 27113629 DOI: 10.1002/humu.23007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 04/03/2016] [Indexed: 02/03/2023]
Abstract
Genome-wide association studies (GWAS) have indicated potential to identify heritability of common complex phenotypes, but traditional approaches have limited ability to detect hiding signals because single SNP has weak effect size accounting for only a small fraction of overall phenotypic variations. To improve the power of GWAS, methods have been developed to identify truly associated genes by jointly testing effects of all SNPs. However, equally considering all SNPs within a gene might dilute strong signals of SNPs in real functional categories. Here, we observed a consistent pattern on enrichment of significant SNPs in eight functional categories across six phenotypes, with the highest enrichment in coding and both UTR regions while the lowest enrichment in the intron. Based on the pattern of SNP enrichment in functional categories, we developed a new approach for detecting gene associations on traits (DGAT) by selecting the most significant functional category and then using SNPs within it to assess gene associations. The method was found to be robust in type I error rate on simulated data, and to have mostly higher power in detecting associated genes for three different diseases than other methods. Further analysis indicated ability of the DGAT to detect novel genes. The DGAT is available by http://sparks-lab.org/server/DGAT.
Collapse
Affiliation(s)
- Huiying Zhao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Dale R Nyholt
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
| | - Yuedong Yang
- Institute for Glycomics, Griffith University, Queensland, Australia
| |
Collapse
|
2006
|
Shahid SU, ᅟ S, Cooper JA, Beaney KE, Li K, Rehman A, Humphries SE. Effect of SORT1, APOB and APOE polymorphisms on LDL-C and coronary heart disease in Pakistani subjects and their comparison with Northwick Park Heart Study II. Lipids Health Dis 2016; 15:83. [PMID: 27112212 PMCID: PMC4845441 DOI: 10.1186/s12944-016-0253-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 04/19/2016] [Indexed: 12/26/2022] Open
Abstract
Background Many SNPs have been identified in genes regulating LDL-C metabolism, but whether their influence is similar in subjects from different ethnicities is unclear. Effect of 4 such SNPs on LDL-C and coronary heart disease (CHD) was examined in Pakistani subjects and was compared with middle aged UK men from Northwick Park Heart Study II (NPHSII). Methods One thousand nine hundred sixty-five (1770 non CHD, 195 CHD) UK and 623 (219 non CHD, 404 CHD) Pakistani subjects were enrolled in the study. The SNPs SORT1 rs646776, APOB rs1042031 and APOE rs429358, rs7412 were genotyped by TaqMan/KASPar technique and their gene score was calculated. LDL-C was calculated by Friedewald equation, results were analyzed using SPSS. Results Allele frequencies were significantly different (p = <0.05) between UK and Pakistani subjects. However, the SNPs were associated with LDL-C in both groups. In UK non CHD, UK CHD, Pakistani non CHD and Pakistani CHD respectively, for rs646776, per risk allele increase in LDL-C(mmol/l) was 0.18(0.04), 0.06(0.11), 0.15(0.04) and 0.27(0.06) respectively. For rs1042031, per risk allele increase in LDL-C in four groups was 0.11(0.04), 0.04(0.14), 0.15(0.06) and 0.25(0.09) respectively. For APOE genotypes, compared to Ɛ3, each Ɛ2 decreased LDL-C by 0.11(0.06), 0.07(0.15), 0.20(0.08) and 0.38(0.09), while each Ɛ4 increased LDL-C by 0.43(0.06), 0.39(0.21), 0.19(0.11) and 0.39(0.14) respectively. Overall gene score explained a considerable proportion of sample variance in four groups (3.8 %, 1.26 % 13.7 % and 12.3 %). Gene score in both non-CHD groups was significantly lower than CHD subjects. Conclusions The SNPs show a dose response association with LDL-C levels and risk of CHD in both populations. Electronic supplementary material The online version of this article (doi:10.1186/s12944-016-0253-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Saleem Ullah Shahid
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan.
| | - Shabana ᅟ
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| | - Jackie A Cooper
- Centre for Cardiovascular Genetics, British Heart Foundation Laboratories, University College London, London, WC1E6JF, UK
| | - Katherine E Beaney
- Centre for Cardiovascular Genetics, British Heart Foundation Laboratories, University College London, London, WC1E6JF, UK
| | - Kawah Li
- Centre for Cardiovascular Genetics, British Heart Foundation Laboratories, University College London, London, WC1E6JF, UK
| | - Abdul Rehman
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| | - Stephen Eric Humphries
- Centre for Cardiovascular Genetics, British Heart Foundation Laboratories, University College London, London, WC1E6JF, UK
| |
Collapse
|
2007
|
Targeted exonic sequencing of GWAS loci in the high extremes of the plasma lipids distribution. Atherosclerosis 2016; 250:63-8. [PMID: 27182959 DOI: 10.1016/j.atherosclerosis.2016.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Genome-wide association studies (GWAS) for plasma lipid levels have mapped numerous genomic loci, with each region often containing many protein-coding genes. Targeted re-sequencing of exons is a strategy to pinpoint causal variants and genes. METHODS We performed solution-based hybrid selection of 9008 exons at 939 genes within 95 GWAS loci for plasma lipid levels and sequenced using next-generation sequencing technology individuals with extremely high as well as low to normal levels of low-density lipoprotein cholesterol (LDL-C, n = 311; mean low = 71 mg/dl versus high = 241 mg/dl), triglycerides (TG, n = 308; mean low = 75 mg/dl versus high = 1938 mg/dl), and high-density lipoprotein cholesterol (HDL-C, n = 684; mean low = 32 mg/dl versus high = 102 mg/dl). We identified 15,002 missense, nonsense, or splice site variants with a frequency <5%. We tested whether coding sequence variants, individually or aggregated within a gene, were associated with plasma lipid levels. To replicate findings, we performed sequencing in independent participants (n = 6424). RESULTS Across discovery and replication sequencing, we found 6 variants with significant associations with plasma lipids. Of these, one was a novel association: p.Ser147Asn variant in APOA4 (14.3% frequency, TG OR = 0.49, P = 7.1 × 10(-4)) with TG. In gene-level association analyses where rare variants within each gene are collapsed, APOC3 (P = 2.1 × 10(-5)) and LDLR (P = 5.0 × 10(-12)) were associated with plasma lipids. CONCLUSIONS After sequencing genes from 95 GWAS loci in participants with extremely high plasma lipid levels, we identified one new coding variant associated with TG. These results provide insight regarding design of similar sequencing studies with respect to sample size, follow-up, and analysis methodology.
Collapse
|
2008
|
Lamiquiz-Moneo I, Blanco-Torrecilla C, Bea AM, Mateo-Gallego R, Pérez-Calahorra S, Baila-Rueda L, Cenarro A, Civeira F, de Castro-Orós I. Frequency of rare mutations and common genetic variations in severe hypertriglyceridemia in the general population of Spain. Lipids Health Dis 2016; 15:82. [PMID: 27108409 PMCID: PMC4842266 DOI: 10.1186/s12944-016-0251-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/18/2016] [Indexed: 11/10/2022] Open
Abstract
Background Hypertriglyceridemia (HTG) is a common complex metabolic trait that results of the accumulation of relatively common genetic variants in combination with other modifier genes and environmental factors resulting in increased plasma triglyceride (TG) levels. The majority of severe primary hypertriglyceridemias is diagnosed in adulthood and their molecular bases have not been fully defined yet. The prevalence of HTG is highly variable among populations, possibly caused by differences in environmental factors and genetic background. However, the prevalence of very high TG and the frequency of rare mutations causing HTG in a whole non-selected population have not been previously studied. Methods The total of 23,310 subjects over 18 years from a primary care-district in a middle-class area of Zaragoza (Spain) with TG >500 mg/dL were selected to establish HTG prevalence. Those affected of primary HTG were considered for further genetic analisys. The promoters, coding regions and exon-intron boundaries of LPL, LMF1, APOC2, APOA5, APOE and GPIHBP1 genes were sequenced. The frequency of rare variants identified was studied in 90 controls. Results One hundred ninety-four subjects (1.04 %) had HTG and 90 subjects (46.4 %) met the inclusion criteria for primary HTG. In this subgroup, nine patients (12.3 %) were carriers of 7 rare variants in LPL, LMF1, APOA5, GPIHBP1 or APOE genes. Three of these mutations are described for the first time in this work. The presence of a rare pathogenic mutation did not confer a differential phenotype or a higher family history of HTG. Conclusion The prevalence of rare mutations in candidate genes in subjects with primary HTG is low. The low frequency of rare mutations, the absence of a more severe phenotype or the dominant transmission of the HTG would not suggest the use of genetic analysis in the clinical practice in this population. Electronic supplementary material The online version of this article (doi:10.1186/s12944-016-0251-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Itziar Lamiquiz-Moneo
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Avenida Isabel La Católica 1-3, 50009, Zaragoza, Spain.
| | - Cristian Blanco-Torrecilla
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Avenida Isabel La Católica 1-3, 50009, Zaragoza, Spain
| | - Ana M Bea
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Avenida Isabel La Católica 1-3, 50009, Zaragoza, Spain
| | - Rocío Mateo-Gallego
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Avenida Isabel La Católica 1-3, 50009, Zaragoza, Spain
| | - Sofía Pérez-Calahorra
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Avenida Isabel La Católica 1-3, 50009, Zaragoza, Spain
| | - Lucía Baila-Rueda
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Avenida Isabel La Católica 1-3, 50009, Zaragoza, Spain
| | - Ana Cenarro
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Avenida Isabel La Católica 1-3, 50009, Zaragoza, Spain
| | - Fernando Civeira
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Avenida Isabel La Católica 1-3, 50009, Zaragoza, Spain
| | - Isabel de Castro-Orós
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Avenida Isabel La Católica 1-3, 50009, Zaragoza, Spain.,Universidad de Zaragoza, Departamento de Bioquímica, Biología Molecular y Celular, 50009, Zaragoza, Spain
| |
Collapse
|
2009
|
Elosua R, Lluís-Ganella C, Subirana I, Havulinna A, Läll K, Lucas G, Sayols-Baixeras S, Pietilä A, Alver M, Cabrera de León A, Sentí M, Siscovick D, Mellander O, Fischer K, Salomaa V, Marrugat J. Cardiovascular Risk Factors and Ischemic Heart Disease: Is the Confluence of Risk Factors Greater Than the Parts? A Genetic Approach. ACTA ACUST UNITED AC 2016; 9:279-86. [PMID: 27103211 DOI: 10.1161/circgenetics.115.001255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 04/18/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiovascular risk factors tend to aggregate. The biological and predictive value of this aggregation is questioned and genetics could shed light on this debate. Our aims were to reappraise the impact of risk factor confluence on ischemic heart disease (IHD) risk by testing whether genetic risk scores (GRSs) associated with these factors interact on an additive or multiplicative scale, and to determine whether these interactions provide additional value for predicting IHD risk. METHODS AND RESULTS We selected genetic variants associated with blood pressure, body mass index, waist circumference, triglycerides, type-2 diabetes mellitus, high-density lipoprotein and low-density lipoprotein cholesterol, and IHD to create GRSs for each factor. We tested and meta-analyzed the impact of additive (synergy index) and multiplicative (βinteraction) interactions between each GRS pair in 1 case-control (n=6042) and 4 cohort studies (n=17 794) and evaluated the predictive value of these interactions. We observed 2 multiplicative interactions: GRSLDL·GRSTriglycerides (βinteraction=-0.096; SE=0.028) and nonpleiotropic GRSIHD·GRSLDL (βinteraction=0.091; SE=0.028). Inclusion of these interaction terms did not improve predictive capacity. CONCLUSIONS The confluence of low-density lipoprotein cholesterol and triglycerides genetic risk load has an additive effect on IHD risk. The interaction between low-density lipoprotein cholesterol and IHD genetic load is more than multiplicative, supporting the hazardous impact on atherosclerosis progression of the combination of inflammation and increased lipid levels. The capacity of risk factor confluence to improve IHD risk prediction is questionable. Further studies in larger samples are warranted to confirm and expand our results.
Collapse
Affiliation(s)
- Roberto Elosua
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.).
| | - Carla Lluís-Ganella
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Isaac Subirana
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Aki Havulinna
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Kristi Läll
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Gavin Lucas
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Sergi Sayols-Baixeras
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Arto Pietilä
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Maris Alver
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Antonio Cabrera de León
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Mariano Sentí
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - David Siscovick
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Olle Mellander
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Krista Fischer
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Veikko Salomaa
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| | - Jaume Marrugat
- From the Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain (R.E., C.L.-G., I.S., G.L., S.S.-B., J.M.); Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain (I.S.); National Institute for Health and Welfare, Helsinki, Finland (A.H., A.P., V.S.); Estonian Genome Center of Tartu University, Tartu, Estonia (K.L., M.A., K.F.); Research Unit, Nuestra Señora de la Candelaria University Hospital, Santa Cruz de Tenerife, Spain (A.C.d.L.); University of La Laguna, La Laguna, Spain (A.C.d.L.); Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain (M.S.); The New York Academy of Medicine (D.S.); and Skåne University Hospital Clinical Research Center, Malmö, Sweden (O.M.)
| |
Collapse
|
2010
|
Tschapalda K, Zhang YQ, Liu L, Golovnina K, Schlemper T, Eichmann TO, Lal-Nag M, Sreenivasan U, McLenithan J, Ziegler S, Sztalryd C, Lass A, Auld D, Oliver B, Waldmann H, Li Z, Shen M, Boxer MB, Beller M. A Class of Diacylglycerol Acyltransferase 1 Inhibitors Identified by a Combination of Phenotypic High-throughput Screening, Genomics, and Genetics. EBioMedicine 2016; 8:49-59. [PMID: 27428418 PMCID: PMC4919474 DOI: 10.1016/j.ebiom.2016.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/02/2016] [Accepted: 04/12/2016] [Indexed: 12/13/2022] Open
Abstract
Excess lipid storage is an epidemic problem in human populations. Thus, the identification of small molecules to treat or prevent lipid storage-related metabolic complications is of great interest. Here we screened > 320.000 compounds for their ability to prevent a cellular lipid accumulation phenotype. We used fly cells because the multifarious tools available for this organism should facilitate unraveling the mechanism-of-action of active small molecules. Of the several hundred lipid storage inhibitors identified in the primary screen we concentrated on three structurally diverse and potent compound classes active in cells of multiple species (including human) and negligible cytotoxicity. Together with Drosophila in vivo epistasis experiments, RNA-Seq expression profiles suggested that the target of one of the small molecules was diacylglycerol acyltransferase 1 (DGAT1), a key enzyme in the production of triacylglycerols and prominent human drug target. We confirmed this prediction by biochemical and enzymatic activity tests. We identified > 600 potent small molecule inhibitors of cellular lipid storage deposition. RNA-Seq expression profiling discriminated the activity of three lead scaffolds and guided subsequent functional studies. We discovered a class of DGAT1 inhibitors, which is active in fly and mammalian cell lines as well as whole flies.
Obesity and other lipid storage associated diseases are a growing health threat of human populations. In an undirected phenotypic screen, we identified pharmacologically active small molecules that reduce or enhance lipid storage. Our work focuses on three lead structures that prevent lipid storage in diverse cellular systems including cells from a diabetes patient. In order to elucidate the compound mechanisms-of-action and cellular targets, we used a combination of RNA-Seq transcriptional profiling and diverse functional assays. Our results strongly suggest that one of our lead structures represents a class of inhibitors targeting the key lipogenic enzyme diacylglycerol acyltransferase 1.
Collapse
Affiliation(s)
- Kirsten Tschapalda
- Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Germany; Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Germany; Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany; Department of Molecular Developmental Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Ya-Qin Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Li Liu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Kseniya Golovnina
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, USA
| | - Thomas Schlemper
- Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Germany; Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Germany
| | | | - Madhu Lal-Nag
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Urmila Sreenivasan
- Department of Medicine, Division of Endocrinology University of Maryland School of Medicine, USA
| | - John McLenithan
- Department of Medicine, Division of Endocrinology University of Maryland School of Medicine, USA
| | - Slava Ziegler
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Carole Sztalryd
- Department of Medicine, Division of Endocrinology University of Maryland School of Medicine, USA; Baltimore VA Medical Center, VA Research Service, Geriatric Research, Education and Clinical Center (GRECC) and VA Maryland Health Care System, Department of Medicine, Division of Endocrinology University of Maryland School of Medicine, USA
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Austria
| | - Douglas Auld
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Brian Oliver
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, USA
| | - Herbert Waldmann
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Zhuyin Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Matthew B Boxer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Mathias Beller
- Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Germany; Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Germany.
| |
Collapse
|
2011
|
Yu H, Cowan CA. Minireview: Genome Editing of Human Pluripotent Stem Cells for Modeling Metabolic Disease. Mol Endocrinol 2016; 30:575-86. [PMID: 27075706 DOI: 10.1210/me.2015-1290] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The pathophysiology of metabolic diseases such as coronary artery disease, diabetes, and obesity is complex and multifactorial. Developing new strategies to prevent or treat these diseases requires in vitro models with which researchers can extensively study the molecular mechanisms that lead to disease. Human pluripotent stem cells and their differentiated derivatives have the potential to provide an unlimited source of disease-relevant cell types and, when combined with recent advances in genome editing, make the goal of generating functional metabolic disease models, for the first time, consistently attainable. However, this approach still has certain limitations including lack of robust differentiation methods and potential off-target effects. This review describes the current progress in human pluripotent stem cell-based metabolic disease research using genome-editing technology.
Collapse
Affiliation(s)
- Haojie Yu
- Department of Stem Cell and Regenerative Biology (H.Y., C.A.C.), Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138; and Center for Regenerative Medicine (C.A.C.), Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Chad A Cowan
- Department of Stem Cell and Regenerative Biology (H.Y., C.A.C.), Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138; and Center for Regenerative Medicine (C.A.C.), Massachusetts General Hospital, Boston, Massachusetts 02114
| |
Collapse
|
2012
|
Chakravarti A, Turner TN. Revealing rate-limiting steps in complex disease biology: The crucial importance of studying rare, extreme-phenotype families. Bioessays 2016; 38:578-86. [DOI: 10.1002/bies.201500203] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Aravinda Chakravarti
- Center for Complex Disease Genomics; McKusick-Nathans Institute of Genetic Medicine; Johns Hopkins University School of Medicine; Baltimore MD USA
| | - Tychele N. Turner
- Center for Complex Disease Genomics; McKusick-Nathans Institute of Genetic Medicine; Johns Hopkins University School of Medicine; Baltimore MD USA
| |
Collapse
|
2013
|
Porta M, Toppila I, Sandholm N, Hosseini SM, Forsblom C, Hietala K, Borio L, Harjutsalo V, Klein BE, Klein R, Paterson AD, Groop PH. Variation in SLC19A3 and Protection From Microvascular Damage in Type 1 Diabetes. Diabetes 2016; 65:1022-30. [PMID: 26718501 PMCID: PMC4806664 DOI: 10.2337/db15-1247] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/17/2015] [Indexed: 12/19/2022]
Abstract
The risk of long-term diabetes complications is not fully explained by diabetes duration or long-term glycemic exposure, suggesting the involvement of genetic factors. Because thiamine regulates intracellular glucose metabolism and corrects for multiple damaging effects of high glucose, we hypothesized that variants in specific thiamine transporters are associated with risk of severe retinopathy and/or severe nephropathy because they modify an individual's ability to achieve sufficiently high intracellular thiamine levels. We tested 134 single nucleotide polymorphisms (SNPs) in two thiamine transporters (SLC19A2/3) and their transcription factors (SP1/2) for an association with severe retinopathy or nephropathy or their combination in the FinnDiane cohort. Subsequently, the results were examined for replication in the DCCT/EDIC and Wisconsin Epidemiologic Study of Diabetic Retinopathy (WESDR) cohorts. We found two SNPs in strong linkage disequilibrium in the SLC19A3 locus associated with a reduced rate of severe retinopathy and the combined phenotype of severe retinopathy and end-stage renal disease. The association for the combined phenotype reached genome-wide significance in a meta-analysis that included the WESDR cohort. These findings suggest that genetic variations in SLC19A3 play an important role in the pathogenesis of severe diabetic retinopathy and nephropathy and may explain why some individuals with type 1 diabetes are less prone than others to develop microvascular complications.
Collapse
Affiliation(s)
- Massimo Porta
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Iiro Toppila
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland Diabetes and Obesity Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland Diabetes and Obesity Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - S Mohsen Hosseini
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland Diabetes and Obesity Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Kustaa Hietala
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland Department of Ophthalmology, Helsinki University Central Hospital, Helsinki, Finland
| | - Lorenzo Borio
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Valma Harjutsalo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland Diabetes and Obesity Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland National Institute for Health and Welfare, Helsinki, Finland
| | - Barbara E Klein
- Department Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI
| | - Ronald Klein
- Department Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI
| | - Andrew D Paterson
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Canada
| | | | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland Diabetes and Obesity Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | | |
Collapse
|
2014
|
Burgess S, Harshfield E. Mendelian randomization to assess causal effects of blood lipids on coronary heart disease: lessons from the past and applications to the future. Curr Opin Endocrinol Diabetes Obes 2016; 23:124-30. [PMID: 26910273 PMCID: PMC4816855 DOI: 10.1097/med.0000000000000230] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Mendelian randomization is a technique for judging the causal impact of a risk factor on an outcome from observational data using genetic variants. Although evidence from Mendelian randomization for the effects of major lipids and lipoproteins on coronary heart disease (CHD) risk has been around for a relatively long time, new data resources and new methodological approaches have given fresh insight into these relationships. The lessons from these analyses are likely to be highly relevant when it comes to lipidomics and the analyses of lipid subspecies whose biology is less well understood. RECENT FINDINGS Although analyses of low-density lipoprotein cholesterol and lipoprotein(a) are unambiguous as there are genetic variants that associate exclusively with these risk factors and have well understood biology, analyses for triglycerides, and high-density lipoprotein cholesterol (HDL-c) are less clear. For example, a subset of genetic variants having specific associations with HDL-c are not associated with CHD risk, but an allele score including all variants associated with HDL-c does associate with CHD risk. Recently developed methods, such as multivariable Mendelian randomization, Mendelian randomization-Egger, and a weighted median method, suggest that the relationship between HDL-c and CHD risk is null, thus confirming experimental evidence. SUMMARY Robust methods for Mendelian randomization have important utility for understanding the causal relationships between major lipids and CHD risk, and are likely to play an important role in judging the causal relevance of lipid subspecies and other metabolites measured on high-dimensional phenotyping platforms.
Collapse
Affiliation(s)
- Stephen Burgess
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Eric Harshfield
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| |
Collapse
|
2015
|
Abstract
PURPOSE OF REVIEW Meta-analyses of major statin trials have suggested that statin therapy modestly increases the risk of developing diabetes. However, the quality of the data on which these findings are based is not without weaknesses and it has also been unclear whether this effect, if true, is an on-target or off-target effect of statins. RECENT FINDINGS In a major Mendelian randomization study of variants in the HMGCR gene, which encodes the protein through which statins exert their effect, two polymorphisms associated with lower LDL-cholesterol were also associated with higher weight, higher waist circumference, higher glucose and higher diabetes risk. These findings correspond with findings from the statin trials. In addition, new observational studies using a genetic risk score for LDL-cholesterol suggest that other pathways linked to LDL-cholesterol metabolism may also affect diabetes risk. SUMMARY Genetic studies indicate that the observed effect of statins on diabetes risk in trials is highly likely to be a true on-target effect. Although other recent studies have suggested that genetically determined lower LDL-cholesterol may be linked to diabetes risk, further data from both genetic studies and clinical trials of other LDL-cholesterol lowering agents are needed to confirm or refute this.
Collapse
Affiliation(s)
- Daniel I Swerdlow
- aDepartment of Medicine, Imperial College London bInstitute of Cardiovascular Science, University College London cNuffield Department of Population Health, University of Oxford, Oxford, UK
| | | |
Collapse
|
2016
|
Jeff JM, Peloso GM, Do R. What can we learn about lipoprotein metabolism and coronary heart disease from studying rare variants? Curr Opin Lipidol 2016; 27:99-104. [PMID: 26844526 PMCID: PMC4819247 DOI: 10.1097/mol.0000000000000277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Rare variant association studies (RVAS) target the class of genetic variation with frequencies less than 1%. Recently, investigators have used exome sequencing in RVAS to identify rare alleles responsible for Mendelian diseases but have experienced greater difficulty discovering such alleles for complex diseases. In this review, we describe what we have learned about lipoprotein metabolism and coronary heart disease through the conduct of RVAS. RECENT FINDINGS Rare protein-altering genetic variation can provide important insights that are not as easily attainable from common variant association studies. First, RVAS can facilitate gene discovery by identifying novel rare protein-altering variants in specific genes that are associated with disease. Second, rare variant associations can provide supportive evidence for putative drug targets for novel therapies. Finally, rare variants can uncover new pathways and reveal new biologic mechanisms. SUMMARY The field of human genetics has already made tremendous progress in understanding lipoprotein metabolism and the causes of coronary heart disease in the context of rare variants. As next generation sequencing becomes more cost-effective, RVAS with larger sample sizes will be conducted. This will lead to more novel rare variant discoveries and the translation of genomic data into biological knowledge and clinical insights for cardiovascular disease.
Collapse
Affiliation(s)
- Janina M. Jeff
- Charles F. Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Gina M. Peloso
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Ron Do
- Charles F. Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- The Center for Statistical Genetics, Icahn School of Medicine at Mount Sinai, New York, NY
- The Zena and Michael A. Weiner Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
2017
|
Therapeutic Targets of Triglyceride Metabolism as Informed by Human Genetics. Trends Mol Med 2016; 22:328-340. [DOI: 10.1016/j.molmed.2016.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/24/2022]
|
2018
|
Abstract
PURPOSE OF REVIEW Combined hyperlipidemia (CHL) is a complex phenotype that is commonly encountered clinically and is often associated with the expression of early heart disease. The affixed adjective 'familial' gives the impression that the trait is monogenic, like familial hypercholesterolemia. But despite significant efforts, genetic studies have yielded little evidence of single gene determinants of CHL. RECENT FINDINGS Sophisticated linkage studies suggest that individual lipid components of the CHL phenotype - such as elevated LDL and triglyceride - each have several determinants that segregate independently in families. Furthermore, DNA sequencing shows that rare large-effect variants in genes such as LDL receptor (LDLR) and lipoprotein lipase are found in some CHL patients, explaining the elevated LDL cholesterol and triglyceride components, respectively. In addition, multiple common small-effect lipid-altering variants accumulate in an individual's genome, raising the LDL cholesterol and/or triglyceride components by multiple mechanisms. Finally, secondary factors, such as poor diet, obesity,fatty liver or diabetes further modulate the expression of the biochemically defined CHL phenotype. SUMMARY Given the current state of genetic understanding, CHL may be best conceptualized as a syndrome with common clinical presentation but multigenic causes, similar to other common conditions such as type 2 diabetes.
Collapse
Affiliation(s)
- Amanda J Brahm
- Department of Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | | |
Collapse
|
2019
|
Ali A, Varga TV, Stojkovic IA, Schulz CA, Hallmans G, Barroso I, Poveda A, Renström F, Orho-Melander M, Franks PW. Do Genetic Factors Modify the Relationship Between Obesity and Hypertriglyceridemia? ACTA ACUST UNITED AC 2016; 9:162-71. [PMID: 26865658 DOI: 10.1161/circgenetics.115.001218] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 01/27/2016] [Indexed: 12/11/2022]
Abstract
Background—
Obesity is a major risk factor for dyslipidemia, but this relationship is highly variable. Recently published data from 2 Danish cohorts suggest that genetic factors may underlie some of this variability.
Methods and Results—
We tested whether established triglyceride-associated loci modify the relationship of body mass index (BMI) and triglyceride concentrations in 2 Swedish cohorts (the Gene–Lifestyle Interactions and Complex Traits Involved in Elevated Disease Risk [GLACIER Study; N=4312] and the Malmö Diet and Cancer Study [N=5352]). The genetic loci were amalgamated into a weighted genetic risk score (WGRS
TG
) by summing the triglyceride-elevating alleles (weighted by their established marginal effects) for all loci. Both BMI and the WGRS
TG
were strongly associated with triglyceride concentrations in GLACIER, with each additional BMI unit (kg/m
2
) associated with 2.8% (
P
=8.4×10
–84
) higher triglyceride concentration and each additional WGRS
TG
unit with 2% (
P
=7.6×10
–48
) higher triglyceride concentration. Each unit of the WGRS
TG
was associated with 1.5% higher triglyceride concentrations in normal weight and 2.4% higher concentrations in overweight/obese participants (
P
interaction
=0.056). Meta-analyses of results from the Swedish cohorts yielded a statistically significant WGRS
TG
×BMI interaction effect (
P
interaction
=6.0×10
–4
), which was strengthened by including data from the Danish cohorts (
P
interaction
=6.5×10
–7
). In the meta-analysis of the Swedish cohorts, nominal evidence of a 3-way interaction (WGRS
TG
×BMI×sex) was observed (
P
interaction
=0.03), where the WGRS
TG
×BMI interaction was only statistically significant in females. Using protein–protein interaction network analyses, we identified molecular interactions and pathways elucidating the metabolic relationships between BMI and triglyceride-associated loci.
Conclusions—
Our findings provide evidence that body fatness accentuates the effects of genetic susceptibility variants in hypertriglyceridemia, effects that are most evident in females.
Collapse
Affiliation(s)
- Ashfaq Ali
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| | - Tibor V. Varga
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| | - Ivana A. Stojkovic
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| | - Christina-Alexandra Schulz
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| | - Göran Hallmans
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| | - Inês Barroso
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| | - Alaitz Poveda
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| | - Frida Renström
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| | - Marju Orho-Melander
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| | - Paul W. Franks
- From the Department of Clinical Sciences, Genetic & Molecular Epidemiology Unit (A.A., T.V.V., A.P., F.R., P.W.F.) and Department of Clinical Sciences, Diabetes & Cardiovascular Disease-Genetic Epidemiology (I.A.S., C.-A.S., M.O.-M.), Lund University, Malmö, Sweden; Department of Systems Medicine, Steno Diabetes Center, Gentofte, Denmark (A.A.); Department of Biobank Research (G.H., F.R.) and Department of Public Health & Clinical Medicine (P.W.F.), Umeå University, Umeå, Sweden; Human
| |
Collapse
|
2020
|
Genome-wide significant results identified for plasma apolipoprotein H levels in middle-aged and older adults. Sci Rep 2016; 6:23675. [PMID: 27030319 PMCID: PMC4814826 DOI: 10.1038/srep23675] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/07/2016] [Indexed: 11/09/2022] Open
Abstract
Apolipoprotein H (ApoH) is a multi-functional plasma glycoprotein that has been associated with negative health outcomes. ApoH levels have high heritability. We undertook a genome-wide association study of ApoH levels using the largest sample to date and replicated the results in an independent cohort (total N = 1,255). In the discovery phase, a meta-analysis of two cohorts, the Sydney Memory and Ageing Study (Sydney MAS) and the Older Australian Twins Study (OATS) (n = 942) revealed genome-wide significant results in or near the APOH gene on chromosome 17 (top SNP, rs7211380, p = 1 × 10−11). The results were replicated in an independent cohort, the Hunter Community Study (p < 0.002) (n = 313). Conditional and joint analysis (COJO) confirmed the association of the chromosomal 17 region with ApoH levels. The set of independent SNPs identified by COJO explained 23% of the variance. The relationships between the top SNPs and cardiovascular/lipid/cognition measures and diabetes were assessed in Sydney MAS, with suggestive results observed for diabetes and cognitive performance. However, replication of these results in the smaller OATS cohort was not found. This work provides impetus for future research to better understand the contribution of genetics to ApoH levels and its possible impacts on health.
Collapse
|
2021
|
Kozlitina J, Zhou H, Brown PN, Rohm RJ, Pan Y, Ayanoglu G, Du X, Rimmer E, Reilly DF, Roddy TP, Cully DF, Vogt TF, Blom D, Hoek M. Plasma Levels of Risk-Variant APOL1 Do Not Associate with Renal Disease in a Population-Based Cohort. J Am Soc Nephrol 2016; 27:3204-3219. [PMID: 27005919 DOI: 10.1681/asn.2015101121] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 02/10/2016] [Indexed: 12/12/2022] Open
Abstract
Two common missense variants in APOL1 (G1 and G2) have been definitively linked to CKD in black Americans. However, not all individuals with the renal-risk genotype develop CKD, and little is known about how APOL1 variants drive disease. Given the association of APOL1 with HDL particles, which are cleared by the kidney, differences in the level or quality of mutant APOL1‑HDL particles could be causal for disease and might serve as a useful risk stratification marker. We measured plasma levels of G0 (low risk), G1, and G2 APOL1 in 3450 individuals in the Dallas Heart Study using a liquid chromatography-MS method that enabled quantitation of the different variants. Additionally, we characterized native APOL1‑HDL from donors with no or two APOL1 risk alleles by size-exclusion chromatography and analysis of immunopurified APOL1‑HDL particles. Finally, we identified genetic loci associated with plasma APOL1 levels and tested for APOL1-dependent association with renal function. Although we replicated the previous association between APOL1 variant status and renal function in nondiabetic individuals, levels of circulating APOL1 did not associate with microalbuminuria or GFR. Furthermore, the size or known components of APOL1‑HDL did not consistently differ in subjects with the renal-risk genotype. Genetic association studies implicated variants in loci harboring haptoglobin-related protein (HPR), APOL1, and ubiquitin D (UBD) in the regulation of plasma APOL1 levels, but these variants did not associate with renal function. Collectively, these data demonstrate that the risk of renal disease associated with APOL1 is probably not related to circulating levels of the mutant protein.
Collapse
Affiliation(s)
- Julia Kozlitina
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Haihong Zhou
- Merck Research Laboratories, Kenilworth, New Jersey
| | | | - Rory J Rohm
- Merck Research Laboratories, Kenilworth, New Jersey
| | - Yi Pan
- Merck Research Laboratories, Kenilworth, New Jersey
| | | | - Xiaoyan Du
- Merck Research Laboratories, Kenilworth, New Jersey
| | - Eric Rimmer
- Merck Research Laboratories, Kenilworth, New Jersey
| | | | | | | | | | - Daniel Blom
- Merck Research Laboratories, Kenilworth, New Jersey
| | - Maarten Hoek
- Merck Research Laboratories, Kenilworth, New Jersey
| |
Collapse
|
2022
|
Norby FL, Eryd SA, Niemeijer MN, Rose LM, Smith AV, Yin X, Agarwal SK, Arking DE, Chasman DL, Chen LY, Eijgelsheim M, Engström G, Franco OH, Heeringa J, Hindy G, Hofman A, Lutsey PL, Magnani JW, McManus DD, Orho-Melander M, Pankow JS, Rukh G, Schulz CA, Uitterlinden AG, Albert CM, Benjamin EJ, Gudnason V, Smith JG, Stricker BHC, Alonso A. Association of Lipid-Related Genetic Variants with the Incidence of Atrial Fibrillation: The AFGen Consortium. PLoS One 2016; 11:e0151932. [PMID: 26999784 PMCID: PMC4801208 DOI: 10.1371/journal.pone.0151932] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/07/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Several studies have shown associations between blood lipid levels and the risk of atrial fibrillation (AF). To test the potential effect of blood lipids with AF risk, we assessed whether previously developed lipid gene scores, used as instrumental variables, are associated with the incidence of AF in 7 large cohorts. METHODS We analyzed 64,901 individuals of European ancestry without previous AF at baseline and with lipid gene scores. Lipid-specific gene scores, based on loci significantly associated with lipid levels, were calculated. Additionally, non-pleiotropic gene scores for high-density lipoprotein cholesterol (HDLc) and low-density lipoprotein cholesterol (LDLc) were calculated using SNPs that were only associated with the specific lipid fraction. Cox models were used to estimate the hazard ratio (HR) and 95% confidence intervals (CI) of AF per 1-standard deviation (SD) increase of each lipid gene score. RESULTS During a mean follow-up of 12.0 years, 5434 (8.4%) incident AF cases were identified. After meta-analysis, the HDLc, LDLc, total cholesterol, and triglyceride gene scores were not associated with incidence of AF. Multivariable-adjusted HR (95% CI) were 1.01 (0.98-1.03); 0.98 (0.96-1.01); 0.98 (0.95-1.02); 0.99 (0.97-1.02), respectively. Similarly, non-pleiotropic HDLc and LDLc gene scores showed no association with incident AF: HR (95% CI) = 1.00 (0.97-1.03); 1.01 (0.99-1.04). CONCLUSIONS In this large cohort study of individuals of European ancestry, gene scores for lipid fractions were not associated with incident AF.
Collapse
Affiliation(s)
- Faye L. Norby
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| | | | - Maartje N. Niemeijer
- Department of Epidemiology, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
| | - Lynda M. Rose
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Albert V. Smith
- Icelandic Heart Association, Research Institute, Kopavogur, Iceland
- The University of Iceland, Reykjavik, Iceland
| | - Xiaoyan Yin
- Cardiology and Preventive Medicine Sections, Department of Biostatistics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sunil K. Agarwal
- Icahn School of Medicine, Mount Sinai Heart Center, New York, New York, United States of America
| | - Dan E. Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Daniel L. Chasman
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lin Y. Chen
- Cardiac Arrhythmia Center, Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Mark Eijgelsheim
- Department of Epidemiology, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Oscar H. Franco
- Department of Epidemiology, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
| | - Jan Heeringa
- Department of Epidemiology, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
| | - George Hindy
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jared W. Magnani
- Cardiology and Preventive Medicine Sections, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- The National Heart, Lung and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - David D. McManus
- Departments of Medicine and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | | | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Gull Rukh
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
| | - Christine M. Albert
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Emelia J. Benjamin
- Cardiology and Preventive Medicine Sections, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- The National Heart, Lung and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Vilmundur Gudnason
- Icelandic Heart Association, Research Institute, Kopavogur, Iceland
- The University of Iceland, Reykjavik, Iceland
| | - J. Gustav Smith
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Lund University, Lund, Sweden
| | - Bruno H. C. Stricker
- Department of Epidemiology, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center—University Medical Center, Rotterdam, The Netherlands
- Inspectorate of Health Care, Utrecht, the Netherlands
| | - Alvaro Alonso
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
2023
|
Goedeke L, Wagschal A, Fernández-Hernando C, Näär AM. miRNA regulation of LDL-cholesterol metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2047-2052. [PMID: 26968099 DOI: 10.1016/j.bbalip.2016.03.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/03/2016] [Accepted: 03/06/2016] [Indexed: 12/18/2022]
Abstract
In the past decade, microRNAs (miRNAs) have emerged as key regulators of circulating levels of lipoproteins. Specifically, recent work has uncovered the role of miRNAs in controlling the levels of atherogenic low-density lipoprotein LDL (LDL)-cholesterol by post-transcriptionally regulating genes involved in very low-density lipoprotein (VLDL) secretion, cholesterol biosynthesis, and hepatic LDL receptor (LDLR) expression. Interestingly, several of these miRNAs are located in genomic loci associated with abnormal levels of circulating lipids in humans. These findings reinforce the interest of targeting this subset of non-coding RNAs as potential therapeutic avenues for regulating plasma cholesterol and triglyceride (TAG) levels. In this review, we will discuss how these new miRNAs represent potential pre-disposition factors for cardiovascular disease (CVD), and putative therapeutic targets in patients with cardiometabolic disorders. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.
Collapse
Affiliation(s)
- Leigh Goedeke
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Alexandre Wagschal
- Massachusetts General Hospital Center for Cancer Research, Charlestown, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, 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, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Anders M Näär
- Massachusetts General Hospital Center for Cancer Research, Charlestown, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2024
|
Abstract
Dyslipidemia is a risk factor for atherosclerotic cardiovascular disease (ASCVD). Abundant data indicate that low-density lipoproteins (LDL) are causal for ASCVD; a new class of LDL-lowering medicines, the PCSK9 inhibitors, will address much unmet medical need. Human genetics suggest that triglyceride-rich lipoproteins (TRL) are pro-atherogenic and have pointed to a number of protein regulators of lipoprotein lipase activity that are candidates for therapeutic targeting. Finally, high-density lipoprotein (HDL) cholesterol does not appear to be causally associated with protection from ASCVD, reinforced by the failure of three CETP inhibitors in CV outcome trials, but HDL function remains of interest.
Collapse
Affiliation(s)
- Daniel J Rader
- Departments of Genetics and Medicine and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
2025
|
Wakil SM, Ram R, Muiya NP, Andres E, Mazhar N, Hagos S, Alshahid M, Meyer BF, Morahan G, Dzimiri N. A common variant association study reveals novel susceptibility loci for low HDL-cholesterol levels in ethnic Arabs. Clin Genet 2016; 90:518-525. [PMID: 26879886 DOI: 10.1111/cge.12761] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/08/2016] [Accepted: 02/11/2016] [Indexed: 01/08/2023]
Abstract
The genetic susceptibility to acquiring low high density lipoprotein-cholesterol (LHDLC) levels is not completely elucidated yet. In this study, we performed a common variant association study for harboring this trait in ethnic Arabs. We employed the Affymetrix high-density Axiom Genome-Wide ASI Array (Asian population) providing a coverage of 598,000 single nucleotide variations (SNPs) to genotype 5495 individuals in a two-phase study involving discovery and validation sets of experiments. The rs1800775 [1.31 (1.22-1.42); p = 3.41E-12] in the CETP gene and rs359027 [1.26 (1.16-1.36); p = 2.55E-08] in the LMCD1 gene were significantly associated with LHDLC levels. Furthermore, rs3104435 [1.26 (1.15-1.38); p = 1.19E-06] at the MATN1 locus, rs9835344 [1.16 (1.08-1.26); p = 8.75E-06] in the CNTN6 gene, rs1559997 [1.3 (1.14-1.47); p = 9.48E-06] in the SDS gene and rs1670273 [1.2 (1.1-1.31); p = 4.81E-06] in the DMN/SYNM gene exhibited suggestive association with the disorder. Seven other variants including rs1147169 in the PLCL1 gene, rs10248618 in the DNAH11, rs476155 in the GLIS3, rs7024300 in the ABCA1, intergenic rs10836699, rs11603691 in P2RX3 and rs750134 in CORO1C gene exhibited borderline protective properties. Validation and joint meta-analysis resulted in rs1800775, rs3104435 and rs359027 retaining their predisposing properties, while rs10836699 and rs11603691 showed protective properties. Our data show several predisposing variants across the genome for LHDLC levels in ethnic Arabs.
Collapse
Affiliation(s)
- S M Wakil
- Genetics Department, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - R Ram
- Western Australian Institute for Medical Research, University of Western Australia, Perth, Australia
| | - N P Muiya
- Genetics Department, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - E Andres
- Genetics Department, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - N Mazhar
- Genetics Department, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - S Hagos
- Genetics Department, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - M Alshahid
- King Faisal Heart Institute, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - B F Meyer
- Genetics Department, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - G Morahan
- Western Australian Institute for Medical Research, University of Western Australia, Perth, Australia
| | - N Dzimiri
- Genetics Department, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
2026
|
Guay SP, Légaré C, Brisson D, Mathieu P, Bossé Y, Gaudet D, Bouchard L. Epigenetic and genetic variations at the TNNT1 gene locus are associated with HDL-C levels and coronary artery disease. Epigenomics 2016; 8:359-71. [PMID: 26950807 DOI: 10.2217/epi.15.120] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
AIM To assess whether epigenetic and genetic variations at the TNNT1 gene locus are associated with high-density lipoprotein cholesterol (HDL-C) and coronary artery disease (CAD). Patients, materials & methods: TNNT1 DNA methylation and c.-20G>A polymorphism were genotyped in subjects with and without familial hypercholesterolemia (FH). RESULTS Lower TNNT1 DNA methylation levels were independently associated with lower HDL-C levels and with the TNNT1 c.-20G>A polymorphism. In FH men, carriers of the TNNT1 c.-20G>A polymorphism had lower HDL-C levels and an increased risk of CAD compared with noncarriers. In non-FH men, a higher TNNT1 DNA methylation level was associated with CAD. CONCLUSION These results suggest that TNNT1 genetic and epigenetic variations are associated with HDL-C levels and CAD.
Collapse
Affiliation(s)
- Simon-Pierre Guay
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,ECOGENE-21 & Lipid Clinic, Chicoutimi Hospital, Saguenay, QC G7H 5H6, Canada
| | - Cécilia Légaré
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,ECOGENE-21 & Lipid Clinic, Chicoutimi Hospital, Saguenay, QC G7H 5H6, Canada
| | - Diane Brisson
- ECOGENE-21 & Lipid Clinic, Chicoutimi Hospital, Saguenay, QC G7H 5H6, Canada.,Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Patrick Mathieu
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC G1V 0A6, Canada
| | - Yohan Bossé
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC G1V 0A6, Canada.,Department of Molecular Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Daniel Gaudet
- ECOGENE-21 & Lipid Clinic, Chicoutimi Hospital, Saguenay, QC G7H 5H6, Canada.,Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Luigi Bouchard
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,ECOGENE-21 & Lipid Clinic, Chicoutimi Hospital, Saguenay, QC G7H 5H6, Canada
| |
Collapse
|
2027
|
Tissue-specific regulatory circuits reveal variable modular perturbations across complex diseases. Nat Methods 2016; 13:366-70. [PMID: 26950747 DOI: 10.1038/nmeth.3799] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 01/26/2016] [Indexed: 12/22/2022]
Abstract
Mapping perturbed molecular circuits that underlie complex diseases remains a great challenge. We developed a comprehensive resource of 394 cell type- and tissue-specific gene regulatory networks for human, each specifying the genome-wide connectivity among transcription factors, enhancers, promoters and genes. Integration with 37 genome-wide association studies (GWASs) showed that disease-associated genetic variants--including variants that do not reach genome-wide significance--often perturb regulatory modules that are highly specific to disease-relevant cell types or tissues. Our resource opens the door to systematic analysis of regulatory programs across hundreds of human cell types and tissues (http://regulatorycircuits.org).
Collapse
|
2028
|
Tragante V, Asselbergs FW, Swerdlow DI, Palmer TM, Moore JH, de Bakker PIW, Keating BJ, Holmes MV. Harnessing publicly available genetic data to prioritize lipid modifying therapeutic targets for prevention of coronary heart disease based on dysglycemic risk. Hum Genet 2016; 135:453-467. [PMID: 26946290 PMCID: PMC4835528 DOI: 10.1007/s00439-016-1647-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/07/2016] [Indexed: 01/14/2023]
Abstract
Therapeutic interventions that lower LDL-cholesterol effectively reduce the risk of coronary artery disease (CAD). However, statins, the most widely prescribed LDL-cholesterol lowering drugs, increase diabetes risk. We used genome-wide association study (GWAS) data in the public domain to investigate the relationship of LDL-C and diabetes and identify loci encoding potential drug targets for LDL-cholesterol modification without causing dysglycemia. We obtained summary-level GWAS data for LDL-C from GLGC, glycemic traits from MAGIC, diabetes from DIAGRAM and CAD from CARDIoGRAMplusC4D consortia. Mendelian randomization analyses identified a one standard deviation (SD) increase in LDL-C caused an increased risk of CAD (odds ratio [OR] 1.63 (95 % confidence interval [CI] 1.55, 1.71), which was not influenced by removing SNPs associated with diabetes. LDL-C/CAD-associated SNPs showed consistent effect directions (binomial P = 6.85 × 10−5). Conversely, a 1-SD increase in LDL-C was causally protective of diabetes (OR 0.86; 95 % CI 0.81, 0.91), however LDL-cholesterol/diabetes-associated SNPs did not show consistent effect directions (binomial P = 0.15). HMGCR, our positive control, associated with LDL-C, CAD and a glycemic composite (derived from GWAS meta-analysis of four glycemic traits and diabetes). In contrast, PCSK9, APOB, LPA, CETP, PLG, NPC1L1 and ALDH2 were identified as “druggable” loci that alter LDL-C and risk of CAD without displaying associations with dysglycemia. In conclusion, LDL-C increases the risk of CAD and the relationship is independent of any association of LDL-C with diabetes. Loci that encode targets of emerging LDL-C lowering drugs do not associate with dysglycemia, and this provides provisional evidence that new LDL-C lowering drugs (such as PCSK9 inhibitors) may not influence risk of diabetes.
Collapse
Affiliation(s)
- Vinicius Tragante
- Department of Heart and Lungs, University Medical Center Utrecht, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Heart and Lungs, University Medical Center Utrecht, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands. .,Institute of Cardiovascular Science, University College London, 222 Euston Road, London, NW1 2DA, UK. .,Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands.
| | - Daniel I Swerdlow
- Institute of Cardiovascular Science, University College London, 222 Euston Road, London, NW1 2DA, UK.,Department of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Tom M Palmer
- Department of Mathematics and Statistics, Lancaster University, Lancaster, UK
| | - Jason H Moore
- Department of Biostatistics and Epidemiology, Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, 19104-6021, USA
| | - Paul I W de Bakker
- Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Brendan J Keating
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Michael V Holmes
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA. .,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA. .,Clinical Trials Services Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, Richard Doll Building, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK.
| |
Collapse
|
2029
|
Nair AK, Piaggi P, McLean NA, Kaur M, Kobes S, Knowler WC, Bogardus C, Hanson RL, Baier LJ. Assessment of established HDL-C loci for association with HDL-C levels and type 2 diabetes in Pima Indians. Diabetologia 2016; 59:481-91. [PMID: 26670163 PMCID: PMC4744100 DOI: 10.1007/s00125-015-3835-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/20/2015] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Epidemiological studies in Pima Indians identified elevated levels of HDL-cholesterol (HDL-C) as a protective factor against type 2 diabetes risk in women. We assessed whether HDL-C-associated single-nucleotide polymorphisms (SNPs) also associate with type 2 diabetes in female Pima Indians. METHODS Twenty-one SNPs in established HDL-C loci were initially analysed in 2,675 full-heritage Pima Indians. SNPs shown to associate with HDL-C (12 SNPs) were assessed for association with type 2 diabetes in 7,710 Pima Indians (55.6% female sex). The CETP locus provided the strongest evidence for association with HDL-C and was further interrogated by analysing tag SNPs. RESULTS Twelve of the 21 SNPs analysed had a significant association with HDL-C in Pima Indians; five SNPs representing four loci (CETP, DOCK6, PPP1R3B and ABCA1) reached genome-wide significance. Three SNPs, at CETP, KLF14 and HNF4A, associated with type 2 diabetes only in female participants with the HDL-C-lowering allele increasing diabetes risk (p values: 3.2 × 10(-4) to 7.7 × 10(-5)); the association remained significant even after adjustment for HDL-C. Additional analysis across CETP identified rs6499863 as having the strongest association with type 2 diabetes in female participants (p = 5.0 × 10(-6)) and this association remained independent of the HDL-C association. CONCLUSIONS/INTERPRETATION SNPs at the CETP, HNF4A and KLF14 locus are associated with HDL-C levels and type 2 diabetes (in female participants). However, since HNF4A and KLF14 are established loci for type 2 diabetes, it is unlikely that HDL-C solely mediates these associations.
Collapse
Affiliation(s)
- Anup K Nair
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 445 North 5th Street, Phoenix, AZ, 85004, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 445 North 5th Street, Phoenix, AZ, 85004, USA
| | - Nellie A McLean
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 445 North 5th Street, Phoenix, AZ, 85004, USA
| | - Manmeet Kaur
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 445 North 5th Street, Phoenix, AZ, 85004, USA
| | - Sayuko Kobes
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 445 North 5th Street, Phoenix, AZ, 85004, USA
| | - William C Knowler
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 445 North 5th Street, Phoenix, AZ, 85004, USA
| | - Clifton Bogardus
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 445 North 5th Street, Phoenix, AZ, 85004, USA
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 445 North 5th Street, Phoenix, AZ, 85004, USA
| | - Leslie J Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 445 North 5th Street, Phoenix, AZ, 85004, USA.
| |
Collapse
|
2030
|
Okada Y, Muramatsu T, Suita N, Kanai M, Kawakami E, Iotchkova V, Soranzo N, Inazawa J, Tanaka T. Significant impact of miRNA-target gene networks on genetics of human complex traits. Sci Rep 2016; 6:22223. [PMID: 26927695 PMCID: PMC4772006 DOI: 10.1038/srep22223] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/01/2016] [Indexed: 11/09/2022] Open
Abstract
The impact of microRNA (miRNA) on the genetics of human complex traits, especially in the context of miRNA-target gene networks, has not been fully assessed. Here, we developed a novel analytical method, MIGWAS, to comprehensively evaluate enrichment of genome-wide association study (GWAS) signals in miRNA–target gene networks. We applied the method to the GWAS results of the 18 human complex traits from >1.75 million subjects, and identified significant enrichment in rheumatoid arthritis (RA), kidney function, and adult height (P < 0.05/18 = 0.0028, most significant enrichment in RA with P = 1.7 × 10−4). Interestingly, these results were consistent with current literature-based knowledge of the traits on miRNA obtained through the NCBI PubMed database search (adjusted P = 0.024). Our method provided a list of miRNA and target gene pairs with excess genetic association signals, part of which included drug target genes. We identified a miRNA (miR-4728-5p) that downregulates PADI2, a novel RA risk gene considered as a promising therapeutic target (rs761426, adjusted P = 2.3 × 10−9). Our study indicated the significant impact of miRNA–target gene networks on the genetics of human complex traits, and provided resources which should contribute to drug discovery and nucleic acid medicine.
Collapse
Affiliation(s)
- Yukinori Okada
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Tomoki Muramatsu
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Naomasa Suita
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Advanced Medicinal Research Laboratories, Tsukuba Research Institute, Ono Pharmaceutical CO., LTD., Tsukuba 300-4247, Japan
| | - Masahiro Kanai
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Eiryo Kawakami
- Laboratory for Disease Systems Modeling, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Valentina Iotchkova
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, UK.,Department of Haematology, University of Cambridge, Hills Rd, Cambridge CB2 0AH, UK
| | - Nicole Soranzo
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, UK.,Department of Haematology, University of Cambridge, Hills Rd, Cambridge CB2 0AH, UK
| | - Johji Inazawa
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Bioresource Research Center, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Toshihiro Tanaka
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Bioresource Research Center, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Laboratory for Cardiovascular Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| |
Collapse
|
2031
|
Pilling LC, Atkins JL, Bowman K, Jones SE, Tyrrell J, Beaumont RN, Ruth KS, Tuke MA, Yaghootkar H, Wood AR, Freathy RM, Murray A, Weedon MN, Xue L, Lunetta K, Murabito JM, Harries LW, Robine JM, Brayne C, Kuchel GA, Ferrucci L, Frayling TM, Melzer D. Human longevity is influenced by many genetic variants: evidence from 75,000 UK Biobank participants. Aging (Albany NY) 2016; 8:547-60. [PMID: 27015805 PMCID: PMC4833145 DOI: 10.18632/aging.100930] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/10/2016] [Indexed: 11/25/2022]
Abstract
Variation in human lifespan is 20 to 30% heritable in twins but few genetic variants have been identified. We undertook a Genome Wide Association Study (GWAS) using age at death of parents of middle-aged UK Biobank participants of European decent (n=75,244 with father's and/or mother's data, excluding early deaths). Genetic risk scores for 19 phenotypes (n=777 proven variants) were also tested. In GWAS, a nicotine receptor locus(CHRNA3, previously associated with increased smoking and lung cancer) was associated with fathers' survival. Less common variants requiring further confirmation were also identified. Offspring of longer lived parents had more protective alleles for coronary artery disease, systolic blood pressure, body mass index, cholesterol and triglyceride levels, type-1 diabetes, inflammatory bowel disease and Alzheimer's disease. In candidate analyses, variants in the TOMM40/APOE locus were associated with longevity, but FOXO variants were not. Associations between extreme longevity (mother >=98 years, fathers >=95 years, n=1,339) and disease alleles were similar, with an additional association with HDL cholesterol (p=5.7x10-3). These results support a multiple protective factors model influencing lifespan and longevity (top 1% survival) in humans, with prominent roles for cardiovascular-related pathways. Several of these genetically influenced risks, including blood pressure and tobacco exposure, are potentially modifiable.
Collapse
Affiliation(s)
- Luke C. Pilling
- Epidemiology and Public Health Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Janice L. Atkins
- Epidemiology and Public Health Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Kirsty Bowman
- Epidemiology and Public Health Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Samuel E. Jones
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Jessica Tyrrell
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Robin N. Beaumont
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Katherine S. Ruth
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Marcus A. Tuke
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Hanieh Yaghootkar
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Andrew R. Wood
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Rachel M. Freathy
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Anna Murray
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Michael N. Weedon
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Luting Xue
- School of Public Health, Department of Biostatistics, Boston University, Boston, MA 02215, USA
| | - Kathryn Lunetta
- School of Public Health, Department of Biostatistics, Boston University, Boston, MA 02215, USA
- The Framingham Heart Study, Framingham, MA 01702, USA
| | - Joanne M. Murabito
- The Framingham Heart Study, Framingham, MA 01702, USA
- Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Lorna W. Harries
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Jean-Marie Robine
- Institut National de la Santé et de la Recherche Médicale (INSERM U1198), 34394 Montpellier, France
- Ecole Pratique des Hautes études (EPHE), 75014 Paris, France
| | - Carol Brayne
- Cambridge Institute of Public Health, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SR, UK
| | - George A. Kuchel
- Center on Aging, University of Connecticut, Farmington, CT 06030, USA
| | | | - Timothy M. Frayling
- Genetics of Complex Traits Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - David Melzer
- Epidemiology and Public Health Group, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
- Center on Aging, University of Connecticut, Farmington, CT 06030, USA
| |
Collapse
|
2032
|
Cusanovich DA, Caliskan M, Billstrand C, Michelini K, Chavarria C, De Leon S, Mitrano A, Lewellyn N, Elias JA, Chupp GL, Lang RM, Shah SJ, Decara JM, Gilad Y, Ober C. Integrated analyses of gene expression and genetic association studies in a founder population. Hum Mol Genet 2016; 25:2104-2112. [PMID: 26931462 PMCID: PMC5062579 DOI: 10.1093/hmg/ddw061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 02/21/2016] [Indexed: 12/17/2022] Open
Abstract
Genome-wide association studies (GWASs) have become a standard tool for dissecting genetic contributions to disease risk. However, these studies typically require extraordinarily large sample sizes to be adequately powered. Strategies that incorporate functional information alongside genetic associations have proved successful in increasing GWAS power. Following this paradigm, we present the results of 20 different genetic association studies for quantitative traits related to complex diseases, conducted in the Hutterites of South Dakota. To boost the power of these association studies, we collected RNA-sequencing data from lymphoblastoid cell lines for 431 Hutterite individuals. We then used Sherlock, a tool that integrates GWAS and expression quantitative trait locus (eQTL) data, to identify weak GWAS signals that are also supported by eQTL data. Using this approach, we found novel associations with quantitative phenotypes related to cardiovascular disease, including carotid intima-media thickness, left atrial volume index, monocyte count and serum YKL-40 levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jack A Elias
- Division of Biology and Medicine, Brown University, Providence, RI 02912, USA and
| | - Geoffrey L Chupp
- Pulmonary and Critical Care, Yale School of Medicine, New Haven, CT 06519, USA
| | - Roberto M Lang
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, IL 60637, USA
| | - Sanjiv J Shah
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, IL 60637, USA
| | - Jeanne M Decara
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
2033
|
Affiliation(s)
- Pradeep Natarajan
- From Broad Institute, Cambridge, MA (P.N.); Cardiology Division, Massachusetts General Hospital, Boston (P.N.); Harvard Medical School, Boston, MA (P.N., C.J.O.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (C.J.O.); and Cardiology Section, Boston Veterans Administration Healthcare System, MA (C.J.O.)
| | - Christopher J O'Donnell
- From Broad Institute, Cambridge, MA (P.N.); Cardiology Division, Massachusetts General Hospital, Boston (P.N.); Harvard Medical School, Boston, MA (P.N., C.J.O.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (C.J.O.); and Cardiology Section, Boston Veterans Administration Healthcare System, MA (C.J.O.).
| |
Collapse
|
2034
|
The effect of genetic variation in PCSK9 on the LDL-cholesterol response to statin therapy. THE PHARMACOGENOMICS JOURNAL 2016; 17:204-208. [PMID: 26902539 DOI: 10.1038/tpj.2016.3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/12/2015] [Accepted: 12/23/2015] [Indexed: 01/14/2023]
Abstract
Statins (HMG-CoA reductase inhibitors) lower low-density lipoprotein cholesterol (LDL-C) and prevent cardiovascular disease. However, there is wide individual variation in LDL-C response. Drugs targeting proprotein convertase subtilin/kexin type 9 (PCSK9) lower LDL-C and will be used with statins. PCSK9 mediates the degradation of LDL receptors (LDLRs). Therefore, a greater LDL-C response to statins would be expected in individuals with PCSK9 loss-of-function (LOF) variants because LDLR degradation is reduced. To examine this hypothesis, the effect of 11 PCSK9 functional variants on statin response was determined in 669 African Americans. One LOF variant, rs11591147 (p.R46L) was significantly associated with LDL-C response to statin (P=0.002). In the three carriers, there was a 55.6% greater LDL-C reduction compared with non-carriers. Another functional variant, rs28362261 (p.N425S), was marginally associated with statin response (P=0.0064).The effect of rs11591147 was present in individuals of European ancestry (N=2388, P=0.054). The therapeutic effect of statins may be modified by genetic variation in PCSK9.
Collapse
|
2035
|
Shi W, Wang Q, Choi W, Li J. Mapping and Congenic Dissection of Genetic Loci Contributing to Hyperglycemia and Dyslipidemia in Mice. PLoS One 2016; 11:e0148462. [PMID: 26859786 PMCID: PMC4747551 DOI: 10.1371/journal.pone.0148462] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/18/2016] [Indexed: 01/01/2023] Open
Abstract
Background Patients with dyslipidemia have an increased risk of developing type 2 diabetes, and diabetic patients often have dyslipidemia. Potential genetic connections of fasting plasma glucose with plasma lipid profile were evaluated using hyperlipidemic mice. Methods 225 male F2 mice were generated from BALB/cJ (BALB) and SM/J(SM) Apoe-deficient (Apoe−/−) mice and fed a Western diet for 5 weeks. Fasting plasma glucose and lipid levels of F2 mice were measured before and after 5 weeks of Western diet and quantitative trait locus (QTL) analysis was performed using data collected from these two time points. 144 SNP(single nucleotide polymorphism) markers across the entire genome were typed. Results One major QTL (logarithm of odds ratio (LOD): 6.46) peaked at 12.7 cM on chromosome 9,Bglu16, and 3 suggestive QTLs on chromosomes 15, 18 and X were identified for fasting glucose, and over 10 loci identified for lipid traits. Bglu16 was adjacent to a major QTL, Hdlq17, for high-density lipoprotein (HDL) cholesterol (LOD: 6.31, peak: 19.1 cM). A congenic strain with a donor chromosomal region harboring Bglu16 and Hdlq17 on the Apoe−/− background showed elevations in plasma glucose and HDL levels. Fasting glucose levels were significantly correlated with non-HDL cholesterol and triglyceride levels, especially on the Western diet, but only marginally correlated with HDL levels in F2 mice. Conclusions We have demonstrated a correlative relationship between fasting glucose and plasma lipids in a segregating F2 population under hyperlipidemic conditions, and this correlation is partially due to genetic linkage between the two disorders.
Collapse
Affiliation(s)
- Weibin Shi
- Departments of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, United States of America.,Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Qian Wang
- Departments of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, United States of America
| | - Wonseok Choi
- Departments of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jing Li
- Departments of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
2036
|
A Simple Test of Class-Level Genetic Association Can Reveal Novel Cardiometabolic Trait Loci. PLoS One 2016; 11:e0148218. [PMID: 26859766 PMCID: PMC4747495 DOI: 10.1371/journal.pone.0148218] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 01/14/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Characterizing the genetic determinants of complex diseases can be further augmented by incorporating knowledge of underlying structure or classifications of the genome, such as newly developed mappings of protein-coding genes, epigenetic marks, enhancer elements and non-coding RNAs. METHODS We apply a simple class-level testing framework, termed Genetic Class Association Testing (GenCAT), to identify protein-coding gene association with 14 cardiometabolic (CMD) related traits across 6 publicly available genome wide association (GWA) meta-analysis data resources. GenCAT uses SNP-level meta-analysis test statistics across all SNPs within a class of elements, as well as the size of the class and its unique correlation structure, to determine if the class is statistically meaningful. The novelty of findings is evaluated through investigation of regional signals. A subset of findings are validated using recently updated, larger meta-analysis resources. A simulation study is presented to characterize overall performance with respect to power, control of family-wise error and computational efficiency. All analysis is performed using the GenCAT package, R version 3.2.1. RESULTS We demonstrate that class-level testing complements the common first stage minP approach that involves individual SNP-level testing followed by post-hoc ascribing of statistically significant SNPs to genes and loci. GenCAT suggests 54 protein-coding genes at 41 distinct loci for the 13 CMD traits investigated in the discovery analysis, that are beyond the discoveries of minP alone. An additional application to biological pathways demonstrates flexibility in defining genetic classes. CONCLUSIONS We conclude that it would be prudent to include class-level testing as standard practice in GWA analysis. GenCAT, for example, can be used as a simple, complementary and efficient strategy for class-level testing that leverages existing data resources, requires only summary level data in the form of test statistics, and adds significant value with respect to its potential for identifying multiple novel and clinically relevant trait associations.
Collapse
|
2037
|
Gusev A, Ko A, Shi H, Bhatia G, Chung W, Penninx BWJH, Jansen R, de Geus EJC, Boomsma DI, Wright FA, Sullivan PF, Nikkola E, Alvarez M, Civelek M, Lusis AJ, Lehtimäki T, Raitoharju E, Kähönen M, Seppälä I, Raitakari OT, Kuusisto J, Laakso M, Price AL, Pajukanta P, Pasaniuc B. Integrative approaches for large-scale transcriptome-wide association studies. Nat Genet 2016; 48:245-52. [PMID: 26854917 DOI: 10.1038/ng.3506] [Citation(s) in RCA: 1485] [Impact Index Per Article: 165.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 01/14/2016] [Indexed: 02/07/2023]
Abstract
Many genetic variants influence complex traits by modulating gene expression, thus altering the abundance of one or multiple proteins. Here we introduce a powerful strategy that integrates gene expression measurements with summary association statistics from large-scale genome-wide association studies (GWAS) to identify genes whose cis-regulated expression is associated with complex traits. We leverage expression imputation from genetic data to perform a transcriptome-wide association study (TWAS) to identify significant expression-trait associations. We applied our approaches to expression data from blood and adipose tissue measured in ∼ 3,000 individuals overall. We imputed gene expression into GWAS data from over 900,000 phenotype measurements to identify 69 new genes significantly associated with obesity-related traits (BMI, lipids and height). Many of these genes are associated with relevant phenotypes in the Hybrid Mouse Diversity Panel. Our results showcase the power of integrating genotype, gene expression and phenotype to gain insights into the genetic basis of complex traits.
Collapse
Affiliation(s)
- Alexander Gusev
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Arthur Ko
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Huwenbo Shi
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, California, USA
| | - Gaurav Bhatia
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Wonil Chung
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Brenda W J H Penninx
- Department of Psychiatry, VU University Medical Center, Amsterdam, the Netherlands
| | - Rick Jansen
- Department of Psychiatry, VU University Medical Center, Amsterdam, the Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, VU University, Amsterdam, the Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, VU University, Amsterdam, the Netherlands
| | - Fred A Wright
- Bioinformatics Research Center, Department of Statistics, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Patrick F Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Elina Nikkola
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Mete Civelek
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Aldons J Lusis
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Pirkanmaa Hospital District and University of Tampere School of Medicine, Tampere, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - Olli T Raitakari
- 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
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Alkes L Price
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Bogdan Pasaniuc
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, California, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
2038
|
Klimentidis YC, Arora A. Interaction of Insulin Resistance and Related Genetic Variants With Triglyceride-Associated Genetic Variants. ACTA ACUST UNITED AC 2016; 9:154-61. [PMID: 26850992 DOI: 10.1161/circgenetics.115.001246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/27/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND Several studies suggest that some triglyceride-associated single-nucleotide polymorphisms (SNPs) have pleiotropic and opposite effects on glycemic traits. This potentially implicates them in pathways such as de novo lipogenesis, which is presumably upregulated in the context of insulin resistance. We therefore tested whether the association of triglyceride-associated SNPs with triglyceride levels differs according to one's level of insulin resistance. METHODS AND RESULTS In 3 cohort studies (combined n=12 487), we tested the interaction of established triglyceride-associated SNPs (individually and collectively) with several traits related to insulin resistance, on triglyceride levels. We also tested the interaction of triglyceride SNPs with fasting insulin-associated SNPs, individually and collectively, on triglyceride levels. We find significant interactions of a weighted genetic risk score for triglycerides with insulin resistance on triglyceride levels (Pinteraction=2.73×10(-11) and Pinteraction=2.48×10(-11) for fasting insulin and homeostasis model assessment of insulin resistance, respectively). The association of the triglyceride genetic risk score with triglyceride levels is >60% stronger among those in the highest tertile of homeostasis model assessment of insulin resistance compared with those in the lowest tertile. Individual SNPs contributing to this trend include those in/near GCKR, CILP2, and IRS1, whereas PIGV-NROB2 and LRPAP1 display an opposite trend of interaction. In the pooled data set, we also identify a SNP-by-SNP interaction involving a triglyceride-associated SNP, rs4722551 near MIR148A, with a fasting insulin-associated SNP, rs4865796 in ARL15 (Pinteraction=4.1×10(-5)). CONCLUSIONS Our findings may thus provide genetic evidence for the upregulation of triglyceride levels in insulin-resistant individuals, in addition to identifying specific genetic loci and a SNP-by-SNP interaction implicated in this process.
Collapse
Affiliation(s)
- Yann C Klimentidis
- From the Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson.
| | - Amit Arora
- From the Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson
| |
Collapse
|
2039
|
Vu KN, Ballantyne CM, Hoogeveen RC, Nambi V, Volcik KA, Boerwinkle E, Morrison AC. Causal Role of Alcohol Consumption in an Improved Lipid Profile: The Atherosclerosis Risk in Communities (ARIC) Study. PLoS One 2016; 11:e0148765. [PMID: 26849558 PMCID: PMC4744040 DOI: 10.1371/journal.pone.0148765] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 01/21/2016] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Health benefits of low-to-moderate alcohol consumption may operate through an improved lipid profile. A Mendelian randomization (MR) approach was used to examine whether alcohol consumption causally affects lipid levels. METHODS This analysis involved 10,893 European Americans (EA) from the Atherosclerosis Risk in Communities (ARIC) study. Common and rare variants in alcohol dehydrogenase and acetaldehyde dehydrogenase genes were evaluated for MR assumptions. Five variants, residing in the ADH1B, ADH1C, and ADH4 genes, were selected as genetic instruments and were combined into an unweighted genetic score. Triglycerides (TG), total cholesterol, high-density lipoprotein cholesterol (HDL-c) and its subfractions (HDL2-c and HDL3-c), low-density lipoprotein cholesterol (LDL-c), small dense LDL-c (sdLDL-c), apolipoprotein B (apoB), and lipoprotein (a) (Lp(a)) levels were analyzed. RESULTS Alcohol consumption significantly increased HDL2-c and reduced TG, total cholesterol, LDL-c, sdLDL-c, and apoB levels. For each of these lipids a non-linear trend was observed. Compared to the first quartile of alcohol consumption, the third quartile had a 12.3% lower level of TG (p < 0.001), a 7.71 mg/dL lower level of total cholesterol (p = 0.007), a 10.3% higher level of HDL2-c (p = 0.007), a 6.87 mg/dL lower level of LDL-c (p = 0.012), a 7.4% lower level of sdLDL-c (p = 0.037), and a 3.5% lower level of apoB (p = 0.058, poverall = 0.022). CONCLUSIONS This study supports the causal role of regular low-to-moderate alcohol consumption in increasing HDL2-c, reducing TG, total cholesterol, and LDL-c, and provides evidence for the novel finding that low-to-moderate consumption of alcohol reduces apoB and sdLDL-c levels among EA. However, given the nonlinearity of the effect of alcohol consumption, even within the range of low-to-moderate drinking, increased consumption does not always result in a larger benefit.
Collapse
Affiliation(s)
- Khanh N. Vu
- School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Christie M. Ballantyne
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, Texas, United States of America
- Houston Methodist Debakey Heart and Vascular Center, Houston, Texas, United States of America
| | - Ron C. Hoogeveen
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, Texas, United States of America
- Houston Methodist Debakey Heart and Vascular Center, Houston, Texas, United States of America
| | - Vijay Nambi
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, Texas, United States of America
- Houston Methodist Debakey Heart and Vascular Center, Houston, Texas, United States of America
- Michael E DeBakey Veterans Affairs Hospital, Houston, Texas, United States of America
| | - Kelly A. Volcik
- Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Eric Boerwinkle
- School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- The Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alanna C. Morrison
- School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
2040
|
Amrutkar M, Kern M, Nuñez-Durán E, Ståhlman M, Cansby E, Chursa U, Stenfeldt E, Borén J, Blüher M, Mahlapuu M. Protein kinase STK25 controls lipid partitioning in hepatocytes and correlates with liver fat content in humans. Diabetologia 2016; 59:341-53. [PMID: 26553096 DOI: 10.1007/s00125-015-3801-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/13/2015] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is closely associated with pathological lipid accumulation in the liver, which is suggested to actively contribute to the development of insulin resistance. We recently identified serine/threonine protein kinase 25 (STK25) as a regulator of liver steatosis, whole-body glucose tolerance and insulin sensitivity in a mouse model system. The aim of this study was to assess the role of STK25 in the control of lipid metabolism in human liver. METHODS Intracellular fat deposition, lipid metabolism and insulin sensitivity were studied in immortalised human hepatocytes (IHHs) and HepG2 hepatocellular carcinoma cells in which STK25 was overexpressed or knocked down by small interfering RNA. The association between STK25 mRNA expression in human liver biopsies and hepatic fat content was analysed. RESULTS Overexpression of STK25 in IHH and HepG2 cells enhanced lipid deposition by suppressing β-oxidation and triacylglycerol (TAG) secretion, while increasing lipid synthesis. Conversely, knockdown of STK25 attenuated lipid accumulation by stimulating β-oxidation and TAG secretion, while inhibiting lipid synthesis. Furthermore, TAG hydrolase activity was repressed in hepatocytes overexpressing STK25 and reciprocally increased in cells with STK25 knockdown. Insulin sensitivity was reduced in STK25-overexpressing cells and enhanced in STK25-deficient hepatocytes. We also found a statistically significant positive correlation between STK25 mRNA expression in human liver biopsies and hepatic fat content. CONCLUSIONS/INTERPRETATION Our data suggest that STK25 regulates lipid partitioning in human liver cells by controlling TAG synthesis as well as lipolytic activity and thereby NEFA release from lipid droplets for β-oxidation and TAG secretion. Our findings highlight STK25 as a potential drug target for the prevention and treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Manoj Amrutkar
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Matthias Kern
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Esther Nuñez-Durán
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Marcus Ståhlman
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Emmelie Cansby
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Urszula Chursa
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Elin Stenfeldt
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Margit Mahlapuu
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden.
| |
Collapse
|
2041
|
Lu Y, Day FR, Gustafsson S, Buchkovich ML, Na J, Bataille V, Cousminer DL, Dastani Z, Drong AW, Esko T, Evans DM, Falchi M, Feitosa MF, Ferreira T, Hedman ÅK, Haring R, Hysi PG, Iles MM, Justice AE, Kanoni S, Lagou V, Li R, Li X, Locke A, Lu C, Mägi R, Perry JRB, Pers TH, Qi Q, Sanna M, Schmidt EM, Scott WR, Shungin D, Teumer A, Vinkhuyzen AAE, Walker RW, Westra HJ, Zhang M, Zhang W, Zhao JH, Zhu Z, Afzal U, Ahluwalia TS, Bakker SJL, Bellis C, Bonnefond A, Borodulin K, Buchman AS, Cederholm T, Choh AC, Choi HJ, Curran JE, de Groot LCPGM, De Jager PL, Dhonukshe-Rutten RAM, Enneman AW, Eury E, Evans DS, Forsen T, Friedrich N, Fumeron F, Garcia ME, Gärtner S, Han BG, Havulinna AS, Hayward C, Hernandez D, Hillege H, Ittermann T, Kent JW, Kolcic I, Laatikainen T, Lahti J, Leach IM, Lee CG, Lee JY, Liu T, Liu Y, Lobbens S, Loh M, Lyytikäinen LP, Medina-Gomez C, Michaëlsson K, Nalls MA, Nielson CM, Oozageer L, Pascoe L, Paternoster L, Polašek O, Ripatti S, Sarzynski MA, Shin CS, Narančić NS, Spira D, Srikanth P, Steinhagen-Thiessen E, Sung YJ, Swart KMA, Taittonen L, Tanaka T, et alLu Y, Day FR, Gustafsson S, Buchkovich ML, Na J, Bataille V, Cousminer DL, Dastani Z, Drong AW, Esko T, Evans DM, Falchi M, Feitosa MF, Ferreira T, Hedman ÅK, Haring R, Hysi PG, Iles MM, Justice AE, Kanoni S, Lagou V, Li R, Li X, Locke A, Lu C, Mägi R, Perry JRB, Pers TH, Qi Q, Sanna M, Schmidt EM, Scott WR, Shungin D, Teumer A, Vinkhuyzen AAE, Walker RW, Westra HJ, Zhang M, Zhang W, Zhao JH, Zhu Z, Afzal U, Ahluwalia TS, Bakker SJL, Bellis C, Bonnefond A, Borodulin K, Buchman AS, Cederholm T, Choh AC, Choi HJ, Curran JE, de Groot LCPGM, De Jager PL, Dhonukshe-Rutten RAM, Enneman AW, Eury E, Evans DS, Forsen T, Friedrich N, Fumeron F, Garcia ME, Gärtner S, Han BG, Havulinna AS, Hayward C, Hernandez D, Hillege H, Ittermann T, Kent JW, Kolcic I, Laatikainen T, Lahti J, Leach IM, Lee CG, Lee JY, Liu T, Liu Y, Lobbens S, Loh M, Lyytikäinen LP, Medina-Gomez C, Michaëlsson K, Nalls MA, Nielson CM, Oozageer L, Pascoe L, Paternoster L, Polašek O, Ripatti S, Sarzynski MA, Shin CS, Narančić NS, Spira D, Srikanth P, Steinhagen-Thiessen E, Sung YJ, Swart KMA, Taittonen L, Tanaka T, Tikkanen E, van der Velde N, van Schoor NM, Verweij N, Wright AF, Yu L, Zmuda JM, Eklund N, Forrester T, Grarup N, Jackson AU, Kristiansson K, Kuulasmaa T, Kuusisto J, Lichtner P, Luan J, Mahajan A, Männistö S, Palmer CD, Ried JS, Scott RA, Stancáková A, Wagner PJ, Demirkan A, Döring A, Gudnason V, Kiel DP, Kühnel B, Mangino M, Mcknight B, Menni C, O'Connell JR, Oostra BA, Shuldiner AR, Song K, Vandenput L, van Duijn CM, Vollenweider P, White CC, Boehnke M, Boettcher Y, Cooper RS, Forouhi NG, Gieger C, Grallert H, Hingorani A, Jørgensen T, Jousilahti P, Kivimaki M, Kumari M, Laakso M, Langenberg C, Linneberg A, Luke A, Mckenzie CA, Palotie A, Pedersen O, Peters A, Strauch K, Tayo BO, Wareham NJ, Bennett DA, Bertram L, Blangero J, Blüher M, Bouchard C, Campbell H, Cho NH, Cummings SR, Czerwinski SA, Demuth I, Eckardt R, Eriksson JG, Ferrucci L, Franco OH, Froguel P, Gansevoort RT, Hansen T, Harris TB, Hastie N, Heliövaara M, Hofman A, Jordan JM, Jula A, Kähönen M, Kajantie E, Knekt PB, Koskinen S, Kovacs P, Lehtimäki T, Lind L, Liu Y, Orwoll ES, Osmond C, Perola M, Pérusse L, Raitakari OT, Rankinen T, Rao DC, Rice TK, Rivadeneira F, Rudan I, Salomaa V, Sørensen TIA, Stumvoll M, Tönjes A, Towne B, Tranah GJ, Tremblay A, Uitterlinden AG, van der Harst P, Vartiainen E, Viikari JS, Vitart V, Vohl MC, Völzke H, Walker M, Wallaschofski H, Wild S, Wilson JF, Yengo L, Bishop DT, Borecki IB, Chambers JC, Cupples LA, Dehghan A, Deloukas P, Fatemifar G, Fox C, Furey TS, Franke L, Han J, Hunter DJ, Karjalainen J, Karpe F, Kaplan RC, Kooner JS, McCarthy MI, Murabito JM, Morris AP, Bishop JAN, North KE, Ohlsson C, Ong KK, Prokopenko I, Richards JB, Schadt EE, Spector TD, Widén E, Willer CJ, Yang J, Ingelsson E, Mohlke KL, Hirschhorn JN, Pospisilik JA, Zillikens MC, Lindgren C, Kilpeläinen TO, Loos RJF. New loci for body fat percentage reveal link between adiposity and cardiometabolic disease risk. Nat Commun 2016; 7:10495. [PMID: 26833246 PMCID: PMC4740398 DOI: 10.1038/ncomms10495] [Show More Authors] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 12/16/2015] [Indexed: 12/24/2022] Open
Abstract
To increase our understanding of the genetic basis of adiposity and its links to cardiometabolic disease risk, we conducted a genome-wide association meta-analysis of body fat percentage (BF%) in up to 100,716 individuals. Twelve loci reached genome-wide significance (P<5 × 10(-8)), of which eight were previously associated with increased overall adiposity (BMI, BF%) and four (in or near COBLL1/GRB14, IGF2BP1, PLA2G6, CRTC1) were novel associations with BF%. Seven loci showed a larger effect on BF% than on BMI, suggestive of a primary association with adiposity, while five loci showed larger effects on BMI than on BF%, suggesting association with both fat and lean mass. In particular, the loci more strongly associated with BF% showed distinct cross-phenotype association signatures with a range of cardiometabolic traits revealing new insights in the link between adiposity and disease risk.
Collapse
Affiliation(s)
- Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, The
Icahn School of Medicine at Mount Sinai, New York, New
York
10029, USA
- The Department of Preventive Medicine, The Icahn School of
Medicine at Mount Sinai, New York, New York
10029, USA
| | - Felix R. Day
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Stefan Gustafsson
- Science for Life Laboratory, Uppsala University, 750
85
Uppsala, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, 751 85
Uppsala, Sweden
| | - Martin L. Buchkovich
- Department of Genetics, University of North Carolina,
Chapel Hill, North Carolina
27599, USA
| | - Jianbo Na
- Department of Developmental and Regenerative Biology, The Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| | - Veronique Bataille
- West Herts NHS Trust, Herts
HP2 4AD, UK
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Diana L. Cousminer
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
| | - Zari Dastani
- Department Epidemiology, Biostatistics and Human Genetics, Lady
Davis Institute, Jewish General Hospital, McGill University,
Montréal, Quebec, Canada
H3T1E2
| | - Alexander W. Drong
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Tõnu Esko
- Estonian Genome Center, Univeristy of Tartu,
Tartu, 51010, Estonia
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachusetts
02115, USA
- Department of Genetics, Harvard Medical School,
Boston, Massachusetts
02115, USA
| | - David M. Evans
- University of Queensland Diamantina Institute, Translational
Research Institute, Brisbane, Queensland
4102, Australia
- MRC Integrative Epidemiology Unit, School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UKnited
| | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
- Department of Genomics of Common Disease, School of Public
Health, Imperial College London, London
W12 0NN, UK
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics,
Washington University School of Medicine, St Louis,
Missouri
63108, USA
| | - Teresa Ferreira
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Åsa K. Hedman
- Science for Life Laboratory, Uppsala University, 750
85
Uppsala, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, 751 85
Uppsala, Sweden
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Robin Haring
- Institute of Clinical Chemistry and Laboratory Medicine,
University Medicine Greifswald, 17475
Greifswald, Germany
- European University of Applied Sciences, Faculty of Applied
Public Health, 18055
Rostock, Germany
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Mark M. Iles
- Leeds Institute of Cancer and Pathology, Cancer Research UK
Leeds Centre, University of Leeds, Leeds
LS9 7TF, UK
| | - Anne E. Justice
- Department of Epidemiology, University of North Carolina at
Chapel Hill, Chapel Hill, North Carolina
27599, USA
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School
of Medicine and Dentistry, Queen Mary University of London,
London
EC1M 6BQ, UK
- Wellcome Trust Sanger Institute, Human Genetics,
Hinxton, Cambridge
CB10 1SA, UK
| | - Vasiliki Lagou
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism,
University of Oxford, Churchill Hospital, Oxford
OX3 7LJ, UK
| | - Rui Li
- Department Epidemiology, Biostatistics and Human Genetics, Lady
Davis Institute, Jewish General Hospital, McGill University,
Montréal, Quebec, Canada
H3T1E2
| | - Xin Li
- Department of Epidemiology, Harvard School of Public
Health, Boston, Massachusetts
02115, USA
| | - Adam Locke
- Center for Statistical Genetics, Department of Biostatistics,
University of Michigan, Ann Arbor, Michigan
48109, USA
| | - Chen Lu
- Department of Biostatistics, Boston University School of Public
Health, Boston, Massachusetts
02118, USA
| | - Reedik Mägi
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Estonian Genome Center, Univeristy of Tartu,
Tartu, 51010, Estonia
| | - John R. B. Perry
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Tune H. Pers
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachusetts
02115, USA
- Department of Genetics, Harvard Medical School,
Boston, Massachusetts
02115, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
- Medical and Population Genetics Program, Broad Institute of MIT
and Harvard, Cambridge
02142, USA
- Department of Epidemiology Research, Statens Serum
Institut, 2100
Copenhagen, Denmark
| | - Qibin Qi
- Department of Epidemiology and Popualtion Health, Albert
Einstein College of Medicine, Bronx, New York
10461, USA
| | - Marianna Sanna
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
- Department of Genomics of Common Disease, School of Public
Health, Imperial College London, London
W12 0NN, UK
| | - Ellen M. Schmidt
- Department of Computational Medicine and Bioinformatics,
University of Michigan, Ann Arbor, Michigan
48109, USA
| | - William R. Scott
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
| | - Dmitry Shungin
- Lund University Diabetes Centre, Department of Clinical
Science, Genetic and Molecular Epidemiology Unit, Skåne University
Hosptial, 205 02
Malmö, Sweden
- Department of Public Health and Clinical Medicine, Unit of
Medicine, Umeå University, 901 87
Umeå, Sweden
- Department of Odontology, Umeå University,
901 85
Umeå, Sweden
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine
Greifswald, 17475
Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics,
University Medicine Greifswald, 17475
Greifswald, Germany
| | | | - Ryan W. Walker
- The Charles Bronfman Institute for Personalized Medicine, The
Icahn School of Medicine at Mount Sinai, New York, New
York
10029, USA
- The Department of Preventive Medicine, The Icahn School of
Medicine at Mount Sinai, New York, New York
10029, USA
| | - Harm-Jan Westra
- Program in Medical and Population Genetics, Broad Institute of
Harvard and Massachusetts Institute of Technology, Cambridge,
Massachusetts
02142, USA
- Divisions of Genetics and Rheumatology, Department of Medicine,
Brigham and Women's Hospital and Harvard Medical School,
Boston, Massachusetts
02446, USA
- Partners Center for Personalized Genetic Medicine,
Boston, Massachusetts
02446, USA
| | - Mingfeng Zhang
- Department of Dermatology, Brigham and Women's
Hospital, Boston, Massachusetts
02115, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Zhihong Zhu
- Queensland Brain Institute, The University of Queensland,
Brisbane
4072, Australia
| | - Uzma Afzal
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
| | - Tarunveer Singh Ahluwalia
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood, Faculty
of Health and Medical Sceinces, University of Copenhagen, 2200
Copenhagen, Denmark
- Danish Pediatric Asthma Center, Gentofte Hospital, The Capital
Region, 2200
Copenhagen, Denmark
- Steno Diabetes Center A/S, DK-2820
Gentofte, Denmark
| | - Stephan J. L. Bakker
- University of Groningen, University Medical Center Groningen,
Department of Medicine, 9700 RB
Groningen, The Netherlands
| | - Claire Bellis
- Department of Genetics, Texas Biomedical Research
Institute, San Antonio, Texas
78245, USA
| | - Amélie Bonnefond
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - Katja Borodulin
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Aron S. Buchman
- Rush Alzheimer's Disease Center, Rush University
Medical Center, Chicago, Illinois
60612, USA
| | - Tommy Cederholm
- Department of Public Health and Caring Sciences, Clinical
Nutrition and Metabolism, Uppsala University, 751 85
Uppsala, Sweden
| | - Audrey C. Choh
- Lifespan Health Research Center, Wright State University
Boonshoft School of Medicine, Dayton, Ohio
45420, USA
| | - Hyung Jin Choi
- Department of Anatomy, Seoul National University College of
Medicine, Seoul
03080, Korea
| | - Joanne E. Curran
- South Texas Diabetes and Obesity Institute, University of Texas
Rio Grande Valley, Brownsville, Texas
78520
| | | | - Philip L. De Jager
- Program in Medical and Population Genetics, Broad Institute of
Harvard and Massachusetts Institute of Technology, Cambridge,
Massachusetts
02142, USA
- Harvard Medical School, Boston,
Massachusetts
02115, USA
- Program in Translational NeuroPsychiatric Genomics, Department
of Neurology, Brigham and Women's Hospital, Boston,
Massachusetts
02115, USA
| | | | - Anke W. Enneman
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Elodie Eury
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute,
San Francisco, California
94107, USA
| | - Tom Forsen
- Department of General Practice and Primary Health Care,
University of Helsinki, FI-00014
Helsinki, Finland
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine,
University Medicine Greifswald, 17475
Greifswald, Germany
| | - Frédéric Fumeron
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers,
F-75006
Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S
1138, Centre de Recherche des Cordeliers, F-75006
Paris, France
- Université Paris Descartes, Sorbonne Paris
Cité, UMR_S 1138, Centre de Recherche des Cordeliers,
F-75006
Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1138,
Centre de Recherche des Cordeliers, F-75006
Paris, France
| | - Melissa E. Garcia
- Laboratory of Epidemiology and Population Sciences, National
Institute on Aging, Bethesda, Maryland
20892, USA
| | - Simone Gärtner
- Department of Medicine A, University Medicine Greifswald,
17475
Greifswald, Germany
| | - Bok-Ghee Han
- Center for Genome Science, National Institute of Health, Osong
Health Technology Administration Complex, Chungcheongbuk-do
370914, Korea
| | - Aki S. Havulinna
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging,
National Institutes of Health, Bethesda, Maryland
20892, USA
| | - Hans Hillege
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
| | - Till Ittermann
- Institute for Community Medicine, University Medicine
Greifswald, 17475
Greifswald, Germany
| | - Jack W. Kent
- Department of Genetics, Texas Biomedical Research
Institute, San Antonio, Texas
78245, USA
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of
Split, Split
21000, Croatia
| | - Tiina Laatikainen
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
- Hospital District of North Karelia, FI-80210
Joensuu, Finland
- Institute of Public Health and Clinical Nutrition, University
of Eastern Finland, FI-70211
Kuopio, Finland
| | - Jari Lahti
- Folkhälsan Research Centre, FI-00290
Helsinki, Finland
- Institute of Behavioural Sciences, University of
Helsinki, FI-00014
Helsinki, Finland
| | - Irene Mateo Leach
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
| | - Christine G. Lee
- Department of Medicine, Oregon Health and Science
University, Portland, Oregon
97239, USA
- Research Service, Veterans Affairs Medical Center,
Portland, Oregon
97239, USA
| | - Jong-Young Lee
- Center for Genome Science, National Institute of Health, Osong
Health Technology Administration Complex, Chungcheongbuk-do
370914, Korea
| | - Tian Liu
- Max Planck Institute for Molecular Genetics, Department of
Vertebrate Genomics, 14195
Berlin, Germany
- Max Planck Institute for Human Development,
14194
Berlin, Germany
| | - Youfang Liu
- Thurston Arthritis Research Center, University of North
Carolina at Chapel Hill, Chaper Hill, North Carolina
27599-7280, USA
| | - Stéphane Lobbens
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - Marie Loh
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Translational Laboratory in Genetic Medicine (TLGM), Agency for
Science, Technology and Research (A*STAR), 8A Biomedical
Grove, Immunos, Level 5, Singapore
138648, Singapore
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, University of Tampere School
of Medicine, FI-33014
Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories and
School of Medicine, University of Tampere, FI-33520
Tampere, Finland
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Karl Michaëlsson
- Department of Surgical Sciences, Orthopedics, Uppsala
University, 751 85
Uppsala, Sweden
| | - Mike A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging,
National Institutes of Health, Bethesda, Maryland
20892, USA
| | - Carrie M. Nielson
- School of Public Health, Oregon Health & Science
University, Portland, Oregon
97239, USA
- Bone & Mineral Unit, Oregon Health & Science
University, Portland, Oregon
97239, USA
| | | | - Laura Pascoe
- Institute of Cell & Molecular Biosciences, Newcastle
University, Newcastle
NE1 7RU, UK
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UKnited
| | - Ozren Polašek
- Department of Public Health, Faculty of Medicine, University of
Split, Split
21000, Croatia
- Centre for Global Health Research, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh, Teviot
Place, Edinburgh
EH8 9AG, UK
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- Wellcome Trust Sanger Institute, Human Genetics,
Hinxton, Cambridge
CB10 1SA, UK
- Hjelt Institute, University of Helsinki,
FI-00014
Helsinki, Finland
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - Chan Soo Shin
- Department of Internal Medicine, Seoul National University
College of Medicine, Seoul
03080, Korea
| | | | - Dominik Spira
- The Berlin Aging Study II; Research Group on Geriatrics;
Charité—Universitätsmedizin Berlin,
13347
Berlin, Germany
- Lipid Clinic at the Interdisciplinary Metabolism Center,
Charité-Universitätsmedizin Berlin, 13353
Berlin, Germany
| | - Priya Srikanth
- School of Public Health, Oregon Health & Science
University, Portland, Oregon
97239, USA
- Bone & Mineral Unit, Oregon Health & Science
University, Portland, Oregon
97239, USA
| | - Elisabeth Steinhagen-Thiessen
- The Berlin Aging Study II; Research Group on Geriatrics;
Charité—Universitätsmedizin Berlin,
13347
Berlin, Germany
- Lipid Clinic at the Interdisciplinary Metabolism Center,
Charité-Universitätsmedizin Berlin, 13353
Berlin, Germany
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of
Medicine, St Louis, Missouri
63110, USA
| | - Karin M. A. Swart
- EMGO Institute for Health and Care Research, VU University
Medical Center, 1081 BT
Amsterdam, The Netherlands
- VUMC, Department of Epidemiology and Biostatistics,
1081 BT
Amsterdam, The Netherlands
| | - Leena Taittonen
- Department of Pediatrics, University of Oulu,
FI-90014
Oulu, Finland
- Department of Pediatrics, Vaasa Central Hospital,
FI-65100
Vaasa, Finland
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on
Aging, Baltimore, Maryland
21225, USA
| | - Emmi Tikkanen
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- Hjelt Institute, University of Helsinki,
FI-00014
Helsinki, Finland
| | - Nathalie van der Velde
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Natasja M. van Schoor
- EMGO Institute for Health and Care Research, VU University
Medical Center, 1081 BT
Amsterdam, The Netherlands
- VUMC, Department of Epidemiology and Biostatistics,
1081 BT
Amsterdam, The Netherlands
| | - Niek Verweij
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University
Medical Center, Chicago, Illinois
60612, USA
| | - Joseph M. Zmuda
- Department of Epidemiology; University of Pittsburgh,
Pittsburgh, Pennsylvania
15261, USA
| | - Niina Eklund
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Terrence Forrester
- Tropical Metabolism Research Unit, Tropical Medicine Research
Institute, University of the West Indies, Mona
JMAAW15, Jamaica
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
| | - Anne U. Jackson
- Center for Statistical Genetics, Department of Biostatistics,
University of Michigan, Ann Arbor, Michigan
48109, USA
| | - Kati Kristiansson
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Teemu Kuulasmaa
- Faculty of Health Sciences, Institute of Clinical Medicine,
Internal Medicine, University of Eastern Finland, 70210
Kuopio, Finland
| | - Johanna Kuusisto
- Faculty of Health Sciences, Institute of Clinical Medicine,
Internal Medicine, University of Eastern Finland, 70210
Kuopio, Finland
- Department of Medicine, University of Eastern Finland,
70210
Kuopio, Finland
- Kuopio University Hospital, 70029
Kuopio, Finland
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Satu Männistö
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Cameron D. Palmer
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachusetts
02115, USA
| | - Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Alena Stancáková
- Department of Medicine, University of Eastern Finland and
Kuopio University Hospital, 70210
Kuopio, Finland
| | - Peter J. Wagner
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Ayse Demirkan
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
University Medical Center, 3015GE
Rotterdam, The Netherlands
| | - Angela Döring
- Institute of Epidemiology I, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur
201, Iceland
- University of Iceland, Faculty of Medicine,
Reykjavik
101, Iceland
| | - Douglas P. Kiel
- Department of Medicine Beth Israel Deaconess Medical Center
and Harvard Medical School, Boston, Massachusetts
02115
- Institute for Aging Research Hebrew Senior Life,
Boston, Massachusetts
02131, USA
| | - Brigitte Kühnel
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Barbara Mcknight
- Cardiovascular Health Research Unit, University of
Washington, Seattle, Washington
98101, USA
- Program in Biostatistics and Biomathematics, Divison of Public
Health Sciences, Fred Hutchinson Cancer Research Center,
Seattle, Washington
98109, USA
- Department of Biostatistics, University of Washington,
Seattle, Washington
98195, USA
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Jeffrey R. O'Connell
- Program for Personalized and Genomic Medicine, Division of
Endocrinology, Diabetes and Nutrition, Department of Medicine, University of
Maryland School of Medicine, Baltimore, Maryland
21201, USA
| | - Ben A. Oostra
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
University Medical Center, 3015GE
Rotterdam, The Netherlands
| | - Alan R. Shuldiner
- Program for Personalized and Genomic Medicine, Division of
Endocrinology, Diabetes and Nutrition, Department of Medicine, University of
Maryland School of Medicine, Baltimore, Maryland
21201, USA
- Geriatric Research and Education Clinical Center, Vetrans
Administration Medical Center, Baltimore, Maryland
21042, USA
| | - Kijoung Song
- Genetics, Projects Clinical Platforms and Sciences,
GlaxoSmithKline, Philadelphia, Pennsylvania
19112, USA
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, 413 45
Gothenburg, Sweden
| | - Cornelia M. van Duijn
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
University Medical Center, 3015GE
Rotterdam, The Netherlands
- Center for Medical Systems Biology, 2300
Leiden, The Netherlands
| | - Peter Vollenweider
- Department of Internal Medicine, University Hospital Lausanne
(CHUV) and University of Lausanne, 1011
Lausanne, Switzerland
| | - Charles C. White
- Department of Biostatistics, Boston University School of Public
Health, Boston, Massachusetts
02118, USA
| | - Michael Boehnke
- Center for Statistical Genetics, Department of Biostatistics,
University of Michigan, Ann Arbor, Michigan
48109, USA
| | - Yvonne Boettcher
- University of Leipzig, IFB Adiposity Diseases,
04103
Leipzig, Germany
- University of Leipzig, Department of Medicine,
04103
Leipzig, Germany
| | - Richard S. Cooper
- Department of Public Health Sciences, Stritch School of
Medicine, Loyola University Chicago, Maywood, Illinois
61053, USA
| | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Harald Grallert
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- German Center for Diabetes Research (DZD),
85764
Neuherberg, Germany
| | - Aroon Hingorani
- Institute of Cardiovascular Science, University College
London, London
WC1E 6BT, UK
| | - Torben Jørgensen
- Department of Clinical Medicine, Faculty of Health and Medical
Sciences, University of Copenhagen, 2200
Copenhagen, Denmark
- Faculty of Medicine, University of Aalborg,
9220
Aalborg, Denmark
- Research Centre for Prevention and Health,
DK2600
Capital Region of Denmark, Denmark
| | - Pekka Jousilahti
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, UCL,
London
WC1E 6BT, UK
| | - Meena Kumari
- Department of Epidemiology and Public Health, UCL,
London
WC1E 6BT, UK
| | - Markku Laakso
- Faculty of Health Sciences, Institute of Clinical Medicine,
Internal Medicine, University of Eastern Finland, 70210
Kuopio, Finland
- Department of Medicine, University of Eastern Finland,
70210
Kuopio, Finland
- Kuopio University Hospital, 70029
Kuopio, Finland
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
- Department of Epidemiology and Public Health, UCL,
London
WC1E 6BT, UK
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup
Hospital, 2600
Glostrup, Denmark
| | - Amy Luke
- Department of Public Health Sciences, Stritch School of
Medicine, Loyola University Chicago, Maywood, Illinois
61053, USA
| | - Colin A. Mckenzie
- Tropical Metabolism Research Unit, Tropical Medicine Research
Institute, University of the West Indies, Mona
JMAAW15, Jamaica
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- Wellcome Trust Sanger Institute, Human Genetics,
Hinxton, Cambridge
CB10 1SA, UK
- Massachusetts General Hospital, Center for Human Genetic
Research, Psychiatric and Neurodevelopmental Genetics Unit,
Boston, Massachusetts
02114, USA
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology,
Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität,
81377
Munich, Germany
| | - Bamidele O. Tayo
- Department of Public Health Sciences, Stritch School of
Medicine, Loyola University Chicago, Maywood, Illinois
61053, USA
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University
Medical Center, Chicago, Illinois
60612, USA
| | - Lars Bertram
- School of Public Health, Faculty of Medicine, Imperial College
London, London
W6 8RP, UK
- Lübeck Interdisciplinary Platform for Genome
Analytics, Institutes of Neurogenetics and Integrative and Experimental
Genomics, University of Lübeck, 23562
Lübeck, Germany
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas
Rio Grande Valley, Brownsville, Texas
78520
| | - Matthias Blüher
- University of Leipzig, IFB Adiposity Diseases,
04103
Leipzig, Germany
- University of Leipzig, Department of Medicine,
04103
Leipzig, Germany
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh, Teviot
Place, Edinburgh
EH8 9AG, UK
| | - Nam H. Cho
- Ajou University School of Medicine, Department of Preventive
Medicine, Suwon Kyoung-gi
443-721, Korea
| | - Steven R. Cummings
- California Pacific Medical Center Research Institute,
San Francisco, California
94107, USA
| | - Stefan A. Czerwinski
- Lifespan Health Research Center, Wright State University
Boonshoft School of Medicine, Dayton, Ohio
45420, USA
| | - Ilja Demuth
- The Berlin Aging Study II; Research Group on Geriatrics;
Charité—Universitätsmedizin Berlin,
13347
Berlin, Germany
- Institute of Medical and Human Genetics,
Charité—Universitätsmedizin Berlin,
13353
Berlin, Germany
| | - Rahel Eckardt
- The Berlin Aging Study II; Research Group on Geriatrics;
Charité—Universitätsmedizin Berlin,
13347
Berlin, Germany
| | - Johan G. Eriksson
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
- Department of General Practice and Primary Health Care,
University of Helsinki, FI-00014
Helsinki, Finland
- Folkhälsan Research Centre, FI-00290
Helsinki, Finland
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on
Aging, Baltimore, Maryland
21225, USA
| | - Oscar H. Franco
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Philippe Froguel
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - Ron T. Gansevoort
- University of Groningen, University Medical Center Groningen,
Department of Medicine, 9700 RB
Groningen, The Netherlands
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern
Denmark, 5000
Odense, Denmark
| | - Tamara B. Harris
- Laboratory of Epidemiology and Population Sciences, National
Institute on Aging, Bethesda, Maryland
20892, USA
| | - Nicholas Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
| | - Markku Heliövaara
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Albert Hofman
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Joanne M. Jordan
- Thurston Arthritis Research Center, University of North
Carolina at Chapel Hill, Chaper Hill, North Carolina
27599-7280, USA
| | - Antti Jula
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University
Hospital, FI-33521
Tampere, Finland
- Department of Clinical Physiology, University of Tampere
School of Medicine, FI-33014
Tampere, Finland
| | - Eero Kajantie
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
- Children's Hospital, Helsinki University Hospital and
University of Helsinki, FI-00029
Helsinki, Finland
- Department of Obstetrics and Gynecology, MRC Oulu, Oulu
University Hospital and University of Oulu, FI-90029
Oulu, Finland
| | - Paul B. Knekt
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Seppo Koskinen
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Peter Kovacs
- University of Leipzig, IFB Adiposity Diseases,
04103
Leipzig, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, University of Tampere School
of Medicine, FI-33014
Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories and
School of Medicine, University of Tampere, FI-33520
Tampere, Finland
| | - Lars Lind
- Department of Medical Sciences, Uppsala University,
751 85
Uppsala, Sweden
| | - Yongmei Liu
- Center for Human Genetics, Division of Public Health Sciences,
Wake Forest School of Medicine, Winston-Salem, North
Carolina
27157, USA
| | - Eric S. Orwoll
- Bone & Mineral Unit, Oregon Health & Science
University, Portland, Oregon
97239, USA
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton,
Southampton General Hospital, Southampton
SO16 6YD, UK
| | - Markus Perola
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- Estonian Genome Center, Univeristy of Tartu,
Tartu, 51010, Estonia
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Louis Pérusse
- Department of Kinesiology, Laval University,
Québec City, Quebec, Canada
G1V 0A6
- Institute of Nutrition and Functional Foods, Laval
University, Québec City, Quebec,
Canada
G1V 0A6
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku
University Hospital, FI-20521
Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular
Medicine, University of Turku, FI-20520
Turku, Finland
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - D. C. Rao
- Division of Statistical Genomics, Department of Genetics,
Washington University School of Medicine, St Louis,
Missouri
63108, USA
- Division of Biostatistics, Washington University School of
Medicine, St Louis, Missouri
63110, USA
- Department of Psychiatry, Washington University School of
Medicine, St Louis, Missouri
63110, USA
| | - Treva K. Rice
- Division of Biostatistics, Washington University School of
Medicine, St Louis, Missouri
63110, USA
- Department of Psychiatry, Washington University School of
Medicine, St Louis, Missouri
63110, USA
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh, Teviot
Place, Edinburgh
EH8 9AG, UK
| | - Veikko Salomaa
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Thorkild I. A. Sørensen
- MRC Integrative Epidemiology Unit, School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UKnited
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg
Hospital, The Capital Region, 2000
Frederiksberg, Denmark
| | - Michael Stumvoll
- University of Leipzig, IFB Adiposity Diseases,
04103
Leipzig, Germany
- University of Leipzig, Department of Medicine,
04103
Leipzig, Germany
| | - Anke Tönjes
- University of Leipzig, Department of Medicine,
04103
Leipzig, Germany
| | - Bradford Towne
- Lifespan Health Research Center, Wright State University
Boonshoft School of Medicine, Dayton, Ohio
45420, USA
| | - Gregory J. Tranah
- California Pacific Medical Center Research Institute,
San Francisco, California
94107, USA
| | - Angelo Tremblay
- Department of Kinesiology, Laval University,
Québec City, Quebec, Canada
G1V 0A6
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity
Cardiology Institute Netherlands-Netherlands Heart Institute, 3501
DG
Utrecht, The Netherlands
- Department of Genetics, University Medical Center Groningen,
University of Groningen, 9700 RB
Groningen, The Netherlands
| | - Erkki Vartiainen
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Jorma S. Viikari
- Department of Medicine, University of Turku,
FI-20521
Turku, Finland
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Laval
University, Québec City, Quebec,
Canada
G1V 0A6
- School of Nutrition, Laval University,
Québec City, Quebec, Canada
G1V 0A6
| | - Henry Völzke
- Institute for Community Medicine, University Medicine
Greifswald, 17475
Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site
Greifswald, 17475
Greifswald, Germany
- DZD (German Centre for Diabetes Research), partner site
Greifswald, 17475
Greifswald, Germany
| | - Mark Walker
- Program in Medical and Population Genetics, Broad Institute of
Harvard and Massachusetts Institute of Technology, Cambridge,
Massachusetts
02142, USA
- Institute of Cellular Medicine, Newcastle University,
Newcastle
NE2 4HH, UK
| | - Henri Wallaschofski
- Institute of Clinical Chemistry and Laboratory Medicine,
University Medicine Greifswald, 17475
Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site
Greifswald, 17475
Greifswald, Germany
| | - Sarah Wild
- Centre for Population Health Sciences, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh,
Edinburgh
EH8 9AG, UK
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
- Centre for Global Health Research, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh, Teviot
Place, Edinburgh
EH8 9AG, UK
| | - Loïc Yengo
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - D. Timothy Bishop
- Leeds Institute of Cancer and Pathology, Cancer Research UK
Leeds Centre, University of Leeds, Leeds
LS9 7TF, UK
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics,
Washington University School of Medicine, St Louis,
Missouri
63108, USA
- Analytical Genetics Group, Regeneron Genetics Center,
Regeneron Pharmaceuticals, Inc., Tarrytown, New York
10591, USA
| | - John C. Chambers
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London
W12 0HS, UK
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public
Health, Boston, Massachusetts
02118, USA
- National Heart, Lung, and Blood Institute, the Framingham
Heart Study, Framingham, Massachusetts
01702, USA
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center,
3000CA
Rotterdam/Zuidholland, The Netherlands
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School
of Medicine and Dentistry, Queen Mary University of London,
London
EC1M 6BQ, UK
- Wellcome Trust Sanger Institute, Human Genetics,
Hinxton, Cambridge
CB10 1SA, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research
of Hereditary Disorders (PACER-HD), King Abdulaziz University,
Jeddah
21589, Saudi Arabia
| | - Ghazaleh Fatemifar
- MRC Integrative Epidemiology Unit, School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UKnited
| | - Caroline Fox
- Harvard Medical School, Boston,
Massachusetts
02115, USA
- National Heart, Lung, and Blood Institute, the Framingham
Heart Study, Framingham, Massachusetts
01702, USA
| | - Terrence S. Furey
- Department of Genetics, University of North Carolina,
Chapel Hill, North Carolina
27599, USA
- Department of Biology, University of North Carolina,
Chapel Hill, North Carolina
27599, USA
| | - Lude Franke
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
- Department of Genetics, University Medical Center Groningen,
University of Groningen, 9700 RB
Groningen, The Netherlands
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of
Public Health, Melvin and Bren Simon Cancer Center,
Indianapolis, Indiana
46202, USA
| | - David J. Hunter
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- Department of Epidemiology, Harvard School of Public
Health, Boston, Massachusetts
02115, USA
- Channing Division of Network Medicine, Department of Medicine,
Brigham and Women's Hospital and Harvard Medical School,
Boston, Massachusetts
02115, USA
- Department of Nutrition, Harvard School of Public
Health, Boston, Massachusetts
02115, USA
| | - Juha Karjalainen
- Department of Genetics, University Medical Center Groningen,
University of Groningen, 9700 RB
Groningen, The Netherlands
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism,
University of Oxford, Churchill Hospital, Oxford
OX3 7LJ, UK
- Oxford NIHR Biomedical Research Centre,
Oxford
OX3 7LJ, UK
| | - Robert C. Kaplan
- Department of Epidemiology and Popualtion Health, Albert
Einstein College of Medicine, Bronx, New York
10461, USA
| | - Jaspal S. Kooner
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London
W12 0HS, UK
- National Heart and Lung Institute, Imperial College
London, London
W12 0NN, UK
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism,
University of Oxford, Churchill Hospital, Oxford
OX3 7LJ, UK
- Oxford NIHR Biomedical Research Centre,
Oxford
OX3 7LJ, UK
| | - Joanne M. Murabito
- Boston University School of Medicine, Department of Medicine,
Section of General Internal Medicine, Boston,
Massachusetts
02118, USA
- NHLBI's and Boston University's Framingham
Heart Study, Framingham, Massachusetts
01702, USA
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Department of Biostatistics, University of Liverpool,
Liverpool
L69 3GA, UK
| | - Julia A. N. Bishop
- Leeds Institute of Cancer and Pathology, Cancer Research UK
Leeds Centre, University of Leeds, Leeds
LS9 7TF, UK
| | - Kari E. North
- Carolina Center for Genome Sciences and Department of
Epidemiology, University of North Carolina at Chapel Hill, Chapel
Hill, North Carolina
27599-7400, USA
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, 413 45
Gothenburg, Sweden
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
- MRC Unit for Lifelong Health and Ageing at UCL,
London
WC1B 5JU, UK
- Department of Paediatrics, University of Cambridge,
Cambridge
CB2 0QQ, UK
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Department of Genomics of Common Disease, School of Public
Health, Imperial College London, London
W12 0NN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism,
University of Oxford, Churchill Hospital, Oxford
OX3 7LJ, UK
| | - J. Brent Richards
- Department Epidemiology, Biostatistics and Human Genetics, Lady
Davis Institute, Jewish General Hospital, McGill University,
Montréal, Quebec, Canada
H3T1E2
- Department of Medicine, Lady Davis Institute, Jewish General
Hospital, McGill University, Montréal,
Quebec, Canada
H3T1E2
- Department of Twin Research, King's College
London, London
SE1 1E7, UK
- Division of Endocrinology, Lady Davis Institute, Jewish
General Hospital, McGill University, Montréal,
Quebec, Canada
H3T1E2
| | - Eric E. Schadt
- Icahn Institute for Genomics and Multiscale Biology, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of
Medicine at Mount Sinai, New York, New York
10029, USA
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
| | - Cristen J. Willer
- Department of Computational Medicine and Bioinformatics,
University of Michigan, Ann Arbor, Michigan
48109, USA
- Department of Human Genetics, University of Michigan,
Ann Arbor, Michigan
48109, USA
- Department of Internal Medicine, Division of Cardiovascular
Medicine, University of Michigan, Ann Arbor, Michigan
48109, USA
| | - Jian Yang
- Queensland Brain Institute, The University of Queensland,
Brisbane
4072, Australia
| | - Erik Ingelsson
- Science for Life Laboratory, Uppsala University, 750
85
Uppsala, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, 751 85
Uppsala, Sweden
- Department of Medicine, Division of Cardiovascular Medicine,
Stanford University School of Medicine, Stanford,
California
94305, USA
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina,
Chapel Hill, North Carolina
27599, USA
| | - Joel N. Hirschhorn
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachusetts
02115, USA
- Department of Genetics, Harvard Medical School,
Boston, Massachusetts
02115, USA
| | - John Andrew Pospisilik
- Department of Epigenetics, Max Planck Institute of
Immunobiology and Epigenetics, D-76108
Freiburg, Germany
| | - M. Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
| | - Cecilia Lindgren
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- The Big Data Institute, University of Oxford,
Oxford
OX3 7LJ, UK
| | - Tuomas Oskari Kilpeläinen
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, The
Icahn School of Medicine at Mount Sinai, New York, New
York
10029, USA
- The Department of Preventive Medicine, The Icahn School of
Medicine at Mount Sinai, New York, New York
10029, USA
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
- The Genetics of Obesity and Related Metabolic Traits Program,
The Icahn School of Medicine at Mount Sinai, New York, New
York, 10029, USA
- The Mindich Child Health and Development Institute, The Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| |
Collapse
|
2042
|
Kim YK, Hwang MY, Kim YJ, Moon S, Han S, Kim BJ. Evaluation of pleiotropic effects among common genetic loci identified for cardio-metabolic traits in a Korean population. Cardiovasc Diabetol 2016; 15:20. [PMID: 26833210 PMCID: PMC4736473 DOI: 10.1186/s12933-016-0337-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/14/2016] [Indexed: 11/10/2022] Open
Abstract
Background The genetic contribution to complex diseases or traits, including cardio-metabolic traits, has been elucidated recently by large-scale genome-wide association studies. These genome-wide association studies have indicated that most pleiotropic loci contain genes associated with lipids. Clinically, lipid related abnormalities are strongly associated with other diseases such as type 2 diabetes, coronary artery disease and hypertension. The aim of this study was to evaluate the shared genetic background of lipids and other cardio-metabolic traits. Methods We conducted meta-analyses of the association between 157 published lipid-associated loci and 10 cardio-metabolic traits in 14,028 Korean individuals genotyped using the Exome chip (Illumina HumanExome BeadChip). We also examined whether the pleiotropic effects of such loci constituted independent (i.e., biological) pleiotropy or mediated pleiotropy in these metabolic pathways. Results Eighteen lipid-associated loci were significantly associated with one of six cardio-metabolic traits after correction for multiple testing (P < 3.70 × 10−4). Region 12q24.12 had pleiotropic effects on fasting plasma glucose, blood pressure and obesity-related traits (body mass index and waist-hip ratio) independent of its effects on the lipid profile. Lipid risk scores, calculated according to whether or not subjects carried the risk allele for lipid traits, were significantly associated with fasting plasma glucose, blood pressure and obesity-related traits. Conclusions The 12q24.12 region showed ethnic-specific genetic pleiotropy among cardio-metabolic traits in this study. Our findings may help to account for molecular mechanisms based on shared genetic background underlying not only dyslipidemia, but also cardiovascular disease and type 2 diabetes. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0337-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yun Kyoung Kim
- Division of Structural and Functional Genomics, Center for Genome Sciences, National Institute of Health, Centers for Disease Control and Prevention, Cheongju-si, Chungcheongbuk-do, 28159, South Korea.
| | - Mi Yeong Hwang
- Division of Structural and Functional Genomics, Center for Genome Sciences, National Institute of Health, Centers for Disease Control and Prevention, Cheongju-si, Chungcheongbuk-do, 28159, South Korea.
| | - Young Jin Kim
- Division of Structural and Functional Genomics, Center for Genome Sciences, National Institute of Health, Centers for Disease Control and Prevention, Cheongju-si, Chungcheongbuk-do, 28159, South Korea.
| | - Sanghoon Moon
- Division of Structural and Functional Genomics, Center for Genome Sciences, National Institute of Health, Centers for Disease Control and Prevention, Cheongju-si, Chungcheongbuk-do, 28159, South Korea.
| | - Sohee Han
- Division of Structural and Functional Genomics, Center for Genome Sciences, National Institute of Health, Centers for Disease Control and Prevention, Cheongju-si, Chungcheongbuk-do, 28159, South Korea.
| | - Bong-Jo Kim
- Division of Structural and Functional Genomics, Center for Genome Sciences, National Institute of Health, Centers for Disease Control and Prevention, Cheongju-si, Chungcheongbuk-do, 28159, South Korea.
| |
Collapse
|
2043
|
Lisik MZ, Gutmajster E, Sieroń AL. Low Levels of HDL in Fragile X Syndrome Patients. Lipids 2016; 51:189-92. [PMID: 26712713 PMCID: PMC4735238 DOI: 10.1007/s11745-015-4109-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/28/2015] [Indexed: 01/08/2023]
Abstract
Fragile X syndrome (FXS) is the most common form of familial mental retardation and one of the leading known causes of autism. The mutation responsible for FXS is a large expansion of the CGG repeats in the promoter region of the FMR1 gene resulting in the transcriptional silencing of the gene in the pathophysiology of Fragile X syndrome was hypothesized. 23 male patients affected by Fragile X syndrome (full mutation in the FMR1 gene) and 24 controls were included in the study. The serum levels of HDL-C were lower in FXS patients (p < 0.001). The serum levels triacylglycerols were higher in FXS patients (p = 0.007) Further study involving larger samples are necessary to confirm the results and define the health implications for abnormal lipid levels in FXS patients.
Collapse
Affiliation(s)
- Małgorzata Z Lisik
- Department of Molecular Biology and Genetics, School of Medicine in Katowice ul., Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland.
| | - Ewa Gutmajster
- Department of Molecular Biology and Genetics, School of Medicine in Katowice ul., Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Aleksander L Sieroń
- Department of Molecular Biology and Genetics, School of Medicine in Katowice ul., Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| |
Collapse
|
2044
|
Kilpeläinen TO, Carli JFM, Skowronski AA, Sun Q, Kriebel J, Feitosa MF, Hedman ÅK, Drong AW, Hayes JE, Zhao J, Pers TH, Schick U, Grarup N, Kutalik Z, Trompet S, Mangino M, Kristiansson K, Beekman M, Lyytikäinen LP, Eriksson J, Henneman P, Lahti J, Tanaka T, Luan J, Greco M FD, Pasko D, Renström F, Willems SM, Mahajan A, Rose LM, Guo X, Liu Y, Kleber ME, Pérusse L, Gaunt T, Ahluwalia TS, Ju Sung Y, Ramos YF, Amin N, Amuzu A, Barroso I, Bellis C, Blangero J, Buckley BM, Böhringer S, I Chen YD, de Craen AJN, Crosslin DR, Dale CE, Dastani Z, Day FR, Deelen J, Delgado GE, Demirkan A, Finucane FM, Ford I, Garcia ME, Gieger C, Gustafsson S, Hallmans G, Hankinson SE, Havulinna AS, Herder C, Hernandez D, Hicks AA, Hunter DJ, Illig T, Ingelsson E, Ioan-Facsinay A, Jansson JO, Jenny NS, Jørgensen ME, Jørgensen T, Karlsson M, Koenig W, Kraft P, Kwekkeboom J, Laatikainen T, Ladwig KH, LeDuc CA, Lowe G, Lu Y, Marques-Vidal P, Meisinger C, Menni C, Morris AP, Myers RH, Männistö S, Nalls MA, Paternoster L, Peters A, Pradhan AD, Rankinen T, Rasmussen-Torvik LJ, Rathmann W, Rice TK, Brent Richards J, Ridker PM, Sattar N, Savage DB, et alKilpeläinen TO, Carli JFM, Skowronski AA, Sun Q, Kriebel J, Feitosa MF, Hedman ÅK, Drong AW, Hayes JE, Zhao J, Pers TH, Schick U, Grarup N, Kutalik Z, Trompet S, Mangino M, Kristiansson K, Beekman M, Lyytikäinen LP, Eriksson J, Henneman P, Lahti J, Tanaka T, Luan J, Greco M FD, Pasko D, Renström F, Willems SM, Mahajan A, Rose LM, Guo X, Liu Y, Kleber ME, Pérusse L, Gaunt T, Ahluwalia TS, Ju Sung Y, Ramos YF, Amin N, Amuzu A, Barroso I, Bellis C, Blangero J, Buckley BM, Böhringer S, I Chen YD, de Craen AJN, Crosslin DR, Dale CE, Dastani Z, Day FR, Deelen J, Delgado GE, Demirkan A, Finucane FM, Ford I, Garcia ME, Gieger C, Gustafsson S, Hallmans G, Hankinson SE, Havulinna AS, Herder C, Hernandez D, Hicks AA, Hunter DJ, Illig T, Ingelsson E, Ioan-Facsinay A, Jansson JO, Jenny NS, Jørgensen ME, Jørgensen T, Karlsson M, Koenig W, Kraft P, Kwekkeboom J, Laatikainen T, Ladwig KH, LeDuc CA, Lowe G, Lu Y, Marques-Vidal P, Meisinger C, Menni C, Morris AP, Myers RH, Männistö S, Nalls MA, Paternoster L, Peters A, Pradhan AD, Rankinen T, Rasmussen-Torvik LJ, Rathmann W, Rice TK, Brent Richards J, Ridker PM, Sattar N, Savage DB, Söderberg S, Timpson NJ, Vandenput L, van Heemst D, Uh HW, Vohl MC, Walker M, Wichmann HE, Widén E, Wood AR, Yao J, Zeller T, Zhang Y, Meulenbelt I, Kloppenburg M, Astrup A, Sørensen TIA, Sarzynski MA, Rao DC, Jousilahti P, Vartiainen E, Hofman A, Rivadeneira F, Uitterlinden AG, Kajantie E, Osmond C, Palotie A, Eriksson JG, Heliövaara M, Knekt PB, Koskinen S, Jula A, Perola M, Huupponen RK, Viikari JS, Kähönen M, Lehtimäki T, Raitakari OT, Mellström D, Lorentzon M, Casas JP, Bandinelli S, März W, Isaacs A, van Dijk KW, van Duijn CM, Harris TB, Bouchard C, Allison MA, Chasman DI, Ohlsson C, Lind L, Scott RA, Langenberg C, Wareham NJ, Ferrucci L, Frayling TM, Pramstaller PP, Borecki IB, Waterworth DM, Bergmann S, Waeber G, Vollenweider P, Vestergaard H, Hansen T, Pedersen O, Hu FB, Eline Slagboom P, Grallert H, Spector TD, Jukema J, Klein RJ, Schadt EE, Franks PW, Lindgren CM, Leibel RL, Loos RJF. Genome-wide meta-analysis uncovers novel loci influencing circulating leptin levels. Nat Commun 2016; 7:10494. [PMID: 26833098 PMCID: PMC4740377 DOI: 10.1038/ncomms10494] [Show More Authors] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 12/16/2015] [Indexed: 01/20/2023] Open
Abstract
Leptin is an adipocyte-secreted hormone, the circulating levels of which correlate closely with overall adiposity. Although rare mutations in the leptin (LEP) gene are well known to cause leptin deficiency and severe obesity, no common loci regulating circulating leptin levels have been uncovered. Therefore, we performed a genome-wide association study (GWAS) of circulating leptin levels from 32,161 individuals and followed up loci reaching P<10(-6) in 19,979 additional individuals. We identify five loci robustly associated (P<5 × 10(-8)) with leptin levels in/near LEP, SLC32A1, GCKR, CCNL1 and FTO. Although the association of the FTO obesity locus with leptin levels is abolished by adjustment for BMI, associations of the four other loci are independent of adiposity. The GCKR locus was found associated with multiple metabolic traits in previous GWAS and the CCNL1 locus with birth weight. Knockdown experiments in mouse adipose tissue explants show convincing evidence for adipogenin, a regulator of adipocyte differentiation, as the novel causal gene in the SLC32A1 locus influencing leptin levels. Our findings provide novel insights into the regulation of leptin production by adipose tissue and open new avenues for examining the influence of variation in leptin levels on adiposity and metabolic health.
Collapse
Affiliation(s)
- Tuomas O. Kilpeläinen
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
- Genetics of Obesity and Related Metabolic Traits Program,
Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine
at Mount Sinai, New York, New York
10029, USA
| | - Jayne F. Martin Carli
- Department of Biochemistry and Molecular Biophysics, Columbia
University, New York, New York
10032, USA
| | - Alicja A. Skowronski
- Institute of Human Nutrition, Columbia University,
New York, New York
10032, USA
| | - Qi Sun
- Channing Division of Network Medicine, Department of Medicine,
Brigham and Women's Hospital and Harvard Medical School,
Boston, Massachussetts
02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public
Health, Boston, Massachussetts
02115, USA
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München - German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
| | - Mary F Feitosa
- Department of Genetics, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | - Åsa K. Hedman
- Science for Life Laboratory, Uppsala University,
Uppsala
750 85, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, Uppsala
751 85, Sweden
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Alexander W. Drong
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - James E. Hayes
- Cell and Developmental Biology Graduate Program, Weill Cornell
Graduate School of Medical Sciences, Cornell University, New
York, New York
10021, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| | - Jinghua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Tune H. Pers
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachussetts
02115, USA
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge, Massachusetts
2142, USA
- Department of Genetics, Harvard Medical School,
Boston, Massachusetts
02115, USA
- Department of Epidemiology Research, Statens Serum
Institut, Copenhagen
2300, Denmark
| | - Ursula Schick
- Genetics of Obesity and Related Metabolic Traits Program,
Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine
at Mount Sinai, New York, New York
10029, USA
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine, Lausanne
University Hospital, Lausanne
1010, Switzerland
- Swiss Institute of Bioinformatics, Lausanne
1015, Switzerland
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University
Medical Center, Leiden
2333, The Netherlands
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
- National Institute for Health Research Biomedical Research
Centre at Guy's and St. Thomas' Foundation Trust,
London
SE1 9RT, UK
| | - Kati Kristiansson
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Institute for Molecular Medicine Finland, University of
Helsinki, Helsinki
FI-00290, Finland
| | - Marian Beekman
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories,
Tampere
FI-33101, Finland
- Department of Clinical Chemistry, University of Tampere School
of Medicine, Tampere
FI-33014, Finland
| | - Joel Eriksson
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Peter Henneman
- Department of Human Genetics, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Department of Clinical Genetics, Amsterdam Medical
Center, Amsterdam
1081 HV, The Netherlands
| | - Jari Lahti
- Institute of Behavioural Sciences, University of
Helsinki, Helsinki
FI-00014, Finland
- Folkhälsan Research Center, Helsinki
FI-00290, Finland
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on
Aging, Baltimore, Maryland
21225, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Fabiola Del Greco M
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC)
- Affiliated Institute of the University of Lübeck,
Bolzano
39100, Italy
| | - Dorota Pasko
- Genetics of Complex Traits, University of Exeter Medical
School, University of Exeter, Exeter
EX2 5DW, UK
| | - Frida Renström
- Department of Clinical Sciences, Genetic and Molecular
Epidemiology Unit, Lund University, Malmö
20502, Sweden
- Department of Biobank Research, Umeå
University, Umeå
90187, Sweden
| | - Sara M. Willems
- Department of Epidemiology, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's
Hospital, Boston, Massachussetts
02215, USA
| | - Xiuqing Guo
- Department of Pediatrics, LABioMed at Harbor-UCLA Medical
Center, Institute for Translational Genomics and Population Sciences,
Torrance, California
90502, USA
| | - Yongmei Liu
- Center for Human Genetics, Division of Public Health Sciences,
Wake Forest School of Medicine, Winston-Salem, North
Carolina
27157, USA
| | - Marcus E. Kleber
- Medical Faculty Mannheim, Vth Department of Medicine,
Heidelberg University, Mannheim
68167, Germany
| | - Louis Pérusse
- Department of Kinesiology, Laval University, Quebec
City, Quebec, Canada
G1V 0A6
- Institute of Nutrition and Functional Foods, Quebec
City, Quebec, Canada
G1V 0A6
| | - Tom Gaunt
- MRC Integrative Epidemiology Unit and School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UK
| | - Tarunveer S. Ahluwalia
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood,
Herlev and Gentofte Hospital, University of Copenhagen, Ledreborg
Allé, Copenhagen
DK-2820, Denmark
- Steno Diabetes Center, Gentofte
DK-2820, Denmark
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of
Medicine, St. Louis, Missouri
63108, USA
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | - Yolande F. Ramos
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
MC, Rotterdam
3015 GE, The Netherlands
| | - Antoinette Amuzu
- Faculty of Epidemiology and Population Health, London School of
Hygiene and Tropical Medicine, London
WC1E 7HT, UK
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Hinxton
CB10 1SA, UK
- NIHR Cambridge Biomedical Research Centre, Institute of
Metabolic Science, Addenbrooke's Hospital, Cambridge
CB2 0QQ, UK
- The University of Cambridge Metabolic Research Laboratories,
Wellcome Trust-MRC Institute of Metabolic Science, Cambridge
CB2 0QQ, UK
| | - Claire Bellis
- Human Genetics, Genome Institute of Singapore, Agency for
Science, Technology and Research of Singapore, Singapore
138672, Singapore
- Genomics Research Centre, Institute of Health and Biomedical
Innovation, Queensland University of Technology, Brisbane,
Queensland
4001, Australia
- Texas Biomedical Research Institute, San
Antonio, Texas
78245, USA
| | - John Blangero
- Texas Biomedical Research Institute, San
Antonio, Texas
78245, USA
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College
Cork, Cork
T12 YT57, Ireland
| | - Stefan Böhringer
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Yii-Der I Chen
- Department of Pediatrics, LABioMed at Harbor-UCLA Medical
Center, Institute for Translational Genomics and Population Sciences,
Torrance, California
90502, USA
| | - Anton J. N. de Craen
- Department of Gerontology and Geriatrics, Leiden University
Medical Center, Leiden
2333, The Netherlands
| | - David R. Crosslin
- Division of Medical Genetics, Department of Medicine,
University of Washington, Seattle, Washington
98195, USA
- Department of Genome Sciences, University of Washington,
Seattle, Washington
98195, USA
| | - Caroline E. Dale
- Faculty of Epidemiology and Population Health, London School of
Hygiene and Tropical Medicine, London
WC1E 7HT, UK
| | - Zari Dastani
- Department of Human Genetics, McGill University,
Montreal, Quebec, Canada
H3A 0G4
| | - Felix R. Day
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Joris Deelen
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Graciela E. Delgado
- Medical Faculty Mannheim, Vth Department of Medicine,
Heidelberg University, Mannheim
68167, Germany
| | - Ayse Demirkan
- Department of Human Genetics, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
MC, Rotterdam
3015 GE, The Netherlands
| | - Francis M. Finucane
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Ian Ford
- Robertson Center for Biostatistics, University of
Glasgow, Glasgow
G12 8QQ, UK
| | - Melissa E. Garcia
- National Heart, Lung, and Blood Institute, NIH,
Bethesda, Maryland
2089, USA
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München - German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München, German Research Center for Environmental Health,
Neuherberg
85764, Germany
| | - Stefan Gustafsson
- Science for Life Laboratory, Uppsala University,
Uppsala
750 85, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, Uppsala
751 85, Sweden
| | - Göran Hallmans
- Department of Biobank Research, Umeå
University, Umeå
90187, Sweden
| | - Susan E. Hankinson
- Channing Division of Network Medicine, Department of Medicine,
Brigham and Women's Hospital and Harvard Medical School,
Boston, Massachussetts
02115, USA
- Department of Biostatistics and Epidemiology, School of Public
Health and Health Sciences, University of Massachusetts,
Amherst, Massachusetts
01003, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public
Health, Boston, Massachusetts
02115, USA
| | - Aki S Havulinna
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Christian Herder
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
- Institute for Clinical Diabetology, German Diabetes Center,
Leibniz Center for Diabetes Research at Heinrich Heine University
Düsseldorf, Düsseldorf
40225, Germany
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging,
Bethesda, Maryland
20892, USA
| | - Andrew A. Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC)
- Affiliated Institute of the University of Lübeck,
Bolzano
39100, Italy
| | - David J. Hunter
- Department of Nutrition and Epidemiology, Harvard T.H. Chan
School of Public Health, Boston, Massachusetts
02115, USA
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München - German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Hannover Unified Biobank, Hannover Medical School,
Hannover
30625, Germany
- Institute for Human Genetics, Hannover Medical School,
Hannover
30625, Germany
| | - Erik Ingelsson
- Science for Life Laboratory, Uppsala University,
Uppsala
750 85, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, Uppsala
751 85, Sweden
- Division of Cardiovascular Medicine, Department of Medicine,
Stanford University School of Medicine, Stanford,
California
94305, USA
| | - Andreea Ioan-Facsinay
- Department of Rheumatology, Leiden University Medical
Center, Leiden
2333, The Netherlands
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and
Physiology, Sahlgrenska Academy, University of Gothenburg,
Gothenburg
41345, Sweden
| | - Nancy S. Jenny
- Laboratory for Clinical Biochemistry Research, Department of
Pathology and Laboratory Medicine, University of Vermont College of
Medicine, Colchester, Vermont
05405, USA
| | | | - Torben Jørgensen
- Research Centre for Prevention and Health, Glostrup University
Hospital, Glostrup
2600, Denmark
- Faculty of Medicine, University of Aalborg,
Aalborg
9100, Denmark
- Faculty of Health and Medical Sciences, University of
Copenhagen, Copenhagen
2200, Denmark
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department
of Clinical Sciences and Orthopaedic Surgery, Lund University, Skåne
University Hospital, Malmö
21428, Sweden
| | - Wolfgang Koenig
- Department of Internal Medicine II - Cardiology, University of
Ulm, Ulm
89081, Germany
- Deutsches Herzzentrum München, Technische
Universität München, Munich
80636, Germany
- DZHK (German Centre for Cardiovascular Research), partner site
Munich Heart Alliance, Munich
80539, Germany
| | - Peter Kraft
- Department of Epidemiology and Biostatistics, Harvard T.H. Chan
School of Public Health, Boston, Massachussetts
02115, USA
| | - Joanneke Kwekkeboom
- Department of Rheumatology, Leiden University Medical
Center, Leiden
2333, The Netherlands
| | - Tiina Laatikainen
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Institute of Public Health and Clinical Nutrition, University
of Eastern Finland, Kuopio
FI-70211, Finland
- Hospital District of North Karelia, Joensuu
FI-80210, Finland
| | - Karl-Heinz Ladwig
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Department of Psychosomatic Medicine and Psychotherapy,
Klinikum Rechts der Isar, Technische Universität
München, Munich
81675, Germany
| | - Charles A. LeDuc
- Division of Molecular Genetics, Department of Pediatrics,
Columbia University, New York, New York
10029, USA
| | - Gordon Lowe
- Institute of Cardiovascular and Medical Sciences, University of
Glasgow, Glasgow
G12 8QQ, UK
| | - Yingchang Lu
- Genetics of Obesity and Related Metabolic Traits Program,
Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine
at Mount Sinai, New York, New York
10029, USA
| | - Pedro Marques-Vidal
- Department of Internal Medicine, Lausanne University
Hospital, Lausanne
1011, Switzerland
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Department of Biostatistics, University of Liverpool,
Liverpool
L69 3GA, UK
| | - Richard H. Myers
- Department of Neurology, Boston University School of
Medicine, Boston, Massachussetts
02118, USA
| | - Satu Männistö
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Mike A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging,
Bethesda, Maryland
20892, USA
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit and School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UK
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
- DZHK (German Centre for Cardiovascular Research), partner site
Munich Heart Alliance, Munich
80539, Germany
| | - Aruna D. Pradhan
- Division of Preventive Medicine, Brigham and Women's
Hospital, Boston, Massachussetts
02215, USA
- Harvard Medical School, Boston,
Massachussetts
02115, USA
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | | | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes
Center, Leibniz Center for Diabetes Research at Heinrich Heine University
Düsseldorf, Düsseldorf
40225, Germany
| | - Treva K. Rice
- Division of Biostatistics, Washington University School of
Medicine, St. Louis, Missouri
63108, USA
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | - J Brent Richards
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
- Department of Medicine, Human Genetics and Epidemiology,
McGill University, Montreal, Quebec, Canada
H3A 0G4
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's
Hospital, Boston, Massachussetts
02215, USA
- Harvard Medical School, Boston,
Massachussetts
02115, USA
| | - Naveed Sattar
- Faculty of Medicine, BHF Glasgow Cardiovascular Research
Centre, Glasgow
G12 8QQ, UK
| | - David B. Savage
- The University of Cambridge Metabolic Research Laboratories,
Wellcome Trust-MRC Institute of Metabolic Science, Cambridge
CB2 0QQ, UK
| | - Stefan Söderberg
- Department of Public Health and Clinical Medicine, Cardiology
and Heart Centre, Umeå University, Umeå
90187, Sweden
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit and School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UK
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Diana van Heemst
- Department of Gerontology and Geriatrics, Leiden University
Medical Center, Leiden
2333, The Netherlands
| | - Hae-Won Uh
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Quebec
City, Quebec, Canada
G1V 0A6
- School of Nutrition, Laval University, Quebec
City, Quebec, Canada
G1V 0A6
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University,
Newcastle upon Tyne
NE1 7RU, UK
| | - Heinz-Erich Wichmann
- Institute of Medical Informatics, Biometry and Epidemiology,
Ludwig-Maximilians-Universität and Klinikum Grosshadern,
Munich
80336, Germany
- Institute of Epidemiology I, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Medical Statistics and Epidemiology, Technical
University Munich, Munich
81675, Germany
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, University of
Helsinki, Helsinki
FI-00290, Finland
| | - Andrew R. Wood
- Genetics of Complex Traits, University of Exeter Medical
School, University of Exeter, Exeter
EX2 5DW, UK
| | - Jie Yao
- Department of Pediatrics, LABioMed at Harbor-UCLA Medical
Center, Institute for Translational Genomics and Population Sciences,
Torrance, California
90502, USA
| | - Tanja Zeller
- German Center for Cardiovascular Research (DZHK e.V.), partner
site Hamburg/Kiel/Lübeck, Hamburg
20246, Germany
- Clinic for General and Interventional Cardiology, University
Heart Center Hamburg, Hamburg
20246, Germany
| | - Yiying Zhang
- Division of Molecular Genetics, Department of Pediatrics,
Columbia University, New York, New York
10029, USA
| | - Ingrid Meulenbelt
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Margreet Kloppenburg
- Department of Rheumatology, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Department of Clinical Epidemiology, Leiden University Medical
Center, Leiden
2333, The Netherlands
| | - Arne Astrup
- Faculty of Science, Department of Nutrition, Exercise, and
Sports, University of Copenhagen, Copenhagen 1165, Denmark
| | - Thorkild I. A. Sørensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- MRC Integrative Epidemiology Unit and School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UK
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg
Hospitals, The Capital Region, Copenhagen
2000, Denmark
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - D. C. Rao
- Department of Genetics, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
- Division of Biostatistics, Washington University School of
Medicine, St. Louis, Missouri
63108, USA
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | - Pekka Jousilahti
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Erkki Vartiainen
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
- Department of Internal Medicine, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
- Department of Internal Medicine, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
| | - Eero Kajantie
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Children's Hospital, Helsinki University Central
Hospital and University of Helsinki, Helsinki
FI-00014, Finland
- Department of Obstetrics and Gynaecology, MRC Oulu, Oulu
University Central Hospital and University of Oulu, Oulu
90220, Finland
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton,
Southampton General Hospital, Southampton
SO16 6YD, UK
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of
Helsinki, Helsinki
FI-00290, Finland
- Wellcome Trust Sanger Institute, Hinxton
CB10 1SA, UK
- Center for Human Genetic Research, Psychiatric and
Neurodevelopmental Genetics Unit, Massachusetts General Hospital,
Boston, Massachusetts
02114, USA
| | - Johan G. Eriksson
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Folkhälsan Research Center, Helsinki
FI-00290, Finland
- Department of General Practice and Primary Health Care,
University of Helsinki, Helsinki
FI-00014, Finland
| | - Markku Heliövaara
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Paul B. Knekt
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Seppo Koskinen
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Antti Jula
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Markus Perola
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Institute for Molecular Medicine Finland, University of
Helsinki, Helsinki
FI-00290, Finland
- University of Tartu, Estonian Genome Centre,
Tartu
51010, Estonia
| | - Risto K. Huupponen
- Department of Pharmacology, Drug Development and Therapeutics,
University of Turku, Turku
FI-20520, Finland
- Unit of Clinical Pharmacology, Turku University
Hospital, Turku
FI-20520, Finland
| | - Jorma S. Viikari
- Division of Medicine, Turku University Hospital,
Turku
FI-20520, Finland
- Department of Medicine, University of Turku,
Turku
FI-20520, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University
Hospital, Tampere
FI-33521, Finland
- Department of Clinical Physiology, University of Tampere
School of Medicine, Tampere
FI-33014, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories,
Tampere
FI-33101, Finland
- Department of Clinical Chemistry, University of Tampere School
of Medicine, Tampere
FI-33014, Finland
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku
University Hospital, Turku
FI-2051, Finland
- Research Centre of Applied and Preventive Cardiovascular
Medicine, University of Turku, Turku
FI-20520, Finland
| | - Dan Mellström
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Mattias Lorentzon
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Juan P. Casas
- Farr Institute of Health Informatics, University College
London, London
NW1 2DA, UK
| | | | - Winfried März
- Medical Faculty Mannheim, Vth Department of Medicine,
Heidelberg University, Mannheim
68167, Germany
- Synlab Academy, Synlab Services LLC, Mannheim
68161, Germany
- Clinical Institute of Medical and Chemical Laboratory
Diagnostics, Medical University of Graz, Graz
8010, Austria
| | - Aaron Isaacs
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
MC, Rotterdam
3015 GE, The Netherlands
| | - Ko W. van Dijk
- Department of Human Genetics, Leiden University Medical
Center, Leiden
2333, The Netherlands
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
MC, Rotterdam
3015 GE, The Netherlands
- Center of Medical Systems Biology, Leiden
2300 RC, The Netherlands
| | - Tamara B. Harris
- Laboratory of Epidemiology and Population Science, National
Institute on Aging, Bethesda, Maryland
20892, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - Matthew A. Allison
- Family and Preventive Medicine, University of
California–San Diego, La Jolla, California
92161, USA
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's
Hospital, Boston, Massachussetts
02215, USA
- Harvard Medical School, Boston,
Massachussetts
02115, USA
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology,
Uppsala University, Uppsala
75185, Sweden
| | - Robert A. Scott
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on
Aging, Baltimore, Maryland
21225, USA
| | - Timothy M. Frayling
- Genetics of Complex Traits, University of Exeter Medical
School, University of Exeter, Exeter
EX2 5DW, UK
| | - Peter P. Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC)
- Affiliated Institute of the University of Lübeck,
Bolzano
39100, Italy
- Department of Neurology, General Central Hospital,
Bolzano
39100, Italy
- Department of Neurology, University of Lübeck,
Lübeck
23562, Germany
| | - Ingrid B. Borecki
- Department of Genetics, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | | | - Sven Bergmann
- Swiss Institute of Bioinformatics, Lausanne
1015, Switzerland
- Department of Medical Genetics, University of Lausanne,
Lausanne
1015, Switzerland
| | - Gérard Waeber
- Department of Internal Medicine, Lausanne University
Hospital, Lausanne
1011, Switzerland
| | - Peter Vollenweider
- Department of Internal Medicine, Lausanne University
Hospital, Lausanne
1011, Switzerland
| | - Henrik Vestergaard
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- Steno Diabetes Center, Gentofte
DK-2820, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- Faculty of Health Sciences, University of Southern
Denmark, Odense
5230, Denmark
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
| | - Frank B. Hu
- Department of Nutrition and Epidemiology, Harvard T.H. Chan
School of Public Health, Boston, Massachusetts
02115, USA
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München - German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - J.W. Jukema
- Department of Cardiology, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands,
Utrecht
3511 EP, The Netherlands
- Durrer Center for Cardiogenetic Research,
Amsterdam
1105 AZ, The Netherlands
| | - Robert J. Klein
- Icahn Institute for Genomics and Multiscale Biology, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| | - Erik E Schadt
- Icahn Institute for Genomics and Multiscale Biology, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| | - Paul W. Franks
- Department of Nutrition, Harvard T.H. Chan School of Public
Health, Boston, Massachussetts
02115, USA
- Department of Clinical Sciences, Genetic and Molecular
Epidemiology Unit, Lund University, Malmö
20502, Sweden
- Department of Public Health and Clinical Medicine,
Umeå University, Umeå
90187, Sweden
| | - Cecilia M. Lindgren
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Program in Medical and Population Genetics, Broad
Institute, Cambridge, Massachussetts
02142, USA
- The Big Data Institute, University of Oxford,
Oxford
OX1 2JD, UK
| | - Rudolph L. Leibel
- Division of Molecular Genetics, Department of Pediatrics,
Columbia University, New York, New York
10029, USA
| | - Ruth J. F. Loos
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
- Genetics of Obesity and Related Metabolic Traits Program,
Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine
at Mount Sinai, New York, New York
10029, USA
- The Mindich Child Health and Development Institute, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| |
Collapse
|
2045
|
Marian AJ. Clinical applications of molecular genetic discoveries. Transl Res 2016; 168:6-14. [PMID: 26548329 PMCID: PMC4718781 DOI: 10.1016/j.trsl.2015.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 10/13/2015] [Accepted: 10/17/2015] [Indexed: 01/08/2023]
Abstract
Genome-wide association studies of complex traits have mapped >15,000 common single nucleotide variants (SNVs). Likewise, applications of massively parallel nucleic acid sequencing technologies often referred to as next-generation sequencing to molecular genetic studies of complex traits have catalogued a large number of rare variants (population frequency of <0.01) in cases with complex traits. Moreover, high-throughput nucleic acid sequencing, variant burden analysis, and linkage studies are illuminating the presence of large number of SNVs in cases and families with single-gene disorders. The plethora of the genetic variants has exposed the formidable challenge of identifying the causal and pathogenic variants from the enormous number of innocuous common and rare variants that exist in the population and in an individual genome. The arduous task of identifying the causal and pathogenic variants is further compounded by the pleiotropic effects of the variants, complexity of cis and trans interactions in the genome, variability in phenotypic expression of the disease, as well as phenotypic plasticity, and the multifarious determinants of the phenotype. Population genetic studies offer the initial roadmaps and have the potential to elucidate novel pathways involved in the pathogenesis of the disease. However, the genome of an individual is unique, rendering unambiguous identification of the causal or pathogenic variant in a single individual exceedingly challenging. Yet, the focus of the practice of medicine is on the individual, as Sir William Osler elegantly expressed in his insightful quotation: "The good physician treats the disease; the great physician treats the patient who has the disease." The daunting task facing physicians, patients, and researchers alike is to apply the modern genetic discoveries to care of the individual with or at risk of the disease.
Collapse
Affiliation(s)
- Ali J Marian
- Center for Cardiovascular Genetics, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Tex; Center for Cardiovascular Genetics, Texas Heart Institute, Houston, Tex.
| |
Collapse
|
2046
|
Loci associated with ischaemic stroke and its subtypes (SiGN): a genome-wide association study. Lancet Neurol 2016; 15:174-184. [PMID: 26708676 PMCID: PMC4912948 DOI: 10.1016/s1474-4422(15)00338-5] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/21/2015] [Accepted: 11/11/2015] [Indexed: 11/21/2022]
Abstract
BACKGROUND The discovery of disease-associated loci through genome-wide association studies (GWAS) is the leading genetic approach to the identification of novel biological pathways underlying diseases in humans. Until recently, GWAS in ischaemic stroke have been limited by small sample sizes and have yielded few loci associated with ischaemic stroke. We did a large-scale GWAS to identify additional susceptibility genes for stroke and its subtypes. METHODS To identify genetic loci associated with ischaemic stroke, we did a two-stage GWAS. In the first stage, we included 16 851 cases with state-of-the-art phenotyping data and 32 473 stroke-free controls. Cases were aged 16 to 104 years, recruited between 1989 and 2012, and subtypes of ischaemic stroke were recorded by centrally trained and certified investigators who used the web-based protocol, Causative Classification of Stroke (CCS). We constructed case-control strata by identifying samples that were genotyped on nearly identical arrays and were of similar genetic ancestral background. We cleaned and imputed data by use of dense imputation reference panels generated from whole-genome sequence data. We did genome-wide testing to identify stroke-associated loci within each stratum for each available phenotype, and we combined summary-level results using inverse variance-weighted fixed-effects meta-analysis. In the second stage, we did in-silico lookups of 1372 single nucleotide polymorphisms identified from the first stage GWAS in 20 941 cases and 364 736 unique stroke-free controls. The ischaemic stroke subtypes of these cases had previously been established with the Trial of Org 10 172 in Acute Stroke Treatment (TOAST) classification system, in accordance with local standards. Results from the two stages were then jointly analysed in a final meta-analysis. FINDINGS We identified a novel locus (G allele at rs12122341) at 1p13.2 near TSPAN2 that was associated with large artery atherosclerosis-related stroke (first stage odds ratio [OR] 1·21, 95% CI 1·13-1·30, p=4·50 × 10-8; joint OR 1·19, 1·12-1·26, p=1·30 × 10-9). Our results also supported robust associations with ischaemic stroke for four other loci that have been reported in previous studies, including PITX2 (first stage OR 1·39, 1·29-1·49, p=3·26 × 10-19; joint OR 1·37, 1·30-1·45, p=2·79 × 10-32) and ZFHX3 (first stage OR 1·19, 1·11-1·27, p=2·93 × 10-7; joint OR 1·17, 1·11-1·23, p=2·29 × 10-10) for cardioembolic stroke, and HDAC9 (first stage OR 1·29, 1·18-1·42, p=3·50 × 10-8; joint OR 1·24, 1·15-1·33, p=4·52 × 10-9) for large artery atherosclerosis stroke. The 12q24 locus near ALDH2, which has previously been associated with all ischaemic stroke but not with any specific subtype, exceeded genome-wide significance in the meta-analysis of small artery stroke (first stage OR 1·20, 1·12-1·28, p=6·82 × 10-8; joint OR 1·17, 1·11-1·23, p=2·92 × 10-9). Other loci associated with stroke in previous studies, including NINJ2, were not confirmed. INTERPRETATION Our results suggest that all ischaemic stroke-related loci previously implicated by GWAS are subtype specific. We identified a novel gene associated with large artery atherosclerosis stroke susceptibility. Follow-up studies will be necessary to establish whether the locus near TSPAN2 can be a target for a novel therapeutic approach to stroke prevention. In view of the subtype-specificity of the associations detected, the rich phenotyping data available in the Stroke Genetics Network (SiGN) are likely to be crucial for further genetic discoveries related to ischaemic stroke. FUNDING US National Institute of Neurological Disorders and Stroke, National Institutes of Health.
Collapse
|
2047
|
Genome-wide association study and targeted metabolomics identifies sex-specific association of CPS1 with coronary artery disease. Nat Commun 2016; 7:10558. [PMID: 26822151 PMCID: PMC4740183 DOI: 10.1038/ncomms10558] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 12/29/2015] [Indexed: 12/21/2022] Open
Abstract
Metabolites derived from dietary choline and L-carnitine, such as trimethylamine N-oxide and betaine, have recently been identified as novel risk factors for atherosclerosis in mice and humans. We sought to identify genetic factors associated with plasma betaine levels and determine their effect on risk of coronary artery disease (CAD). A two-stage genome-wide association study (GWAS) identified two significantly associated loci on chromosomes 2q34 and 5q14.1. The lead variant on 2q24 (rs715) localizes to carbamoyl-phosphate synthase 1 (CPS1), which encodes a mitochondrial enzyme that catalyses the first committed reaction and rate-limiting step in the urea cycle. Rs715 is also significantly associated with decreased levels of urea cycle metabolites and increased plasma glycine levels. Notably, rs715 yield a strikingly significant and protective association with decreased risk of CAD in only women. These results suggest that glycine metabolism and/or the urea cycle represent potentially novel sex-specific mechanisms for the development of atherosclerosis. Dietary choline metabolites, such as trimethylamine N-oxide and betaine, have been associated with coronary artery disease (CAD). Here, Hartiala et al. identify two genetic loci for betaine levels on chromosomes 2q34 and 5q14.1 and find that the 2q34 locus was also associated with other pathway intermediates, and decreased risk of CAD in women.
Collapse
|
2048
|
Abstract
Although relatively rare, pancreatic tumors are highly lethal [1]. In the United States, an estimated 48,960 individuals will be diagnosed with pancreatic cancer and 40,560 will die from this disease in 2015 [1]. Globally, 337,872 new pancreatic cancer cases and 330,391 deaths were estimated in 2012 [2]. In contrast to most other cancers, mortality rates for pancreatic cancer are not improving; in the US, it is predicted to become the second leading cause of cancer related deaths by 2030 [3, 4]. The vast majority of tumors arise in the exocrine pancreas, with pancreatic ductal adenocarcinoma (PDAC) accounting for approximately 95% of tumors. Tumors arising in the endocrine pancreas (pancreatic neuroendocrine tumors) represent less than 5% of all pancreatic tumors [5]. Smoking, type 2 diabetes mellitus (T2D), obesity and pancreatitis are the most consistent epidemiological risk factors for pancreatic cancer [5]. Family history is also a risk factor for developing pancreatic cancer with odds ratios (OR) ranging from 1.7-2.3 for first-degree relatives in most studies, indicating that shared genetic factors may play a role in the etiology of this disease [6-9]. This review summarizes the current knowledge of germline pancreatic cancer risk variants with a special emphasis on common susceptibility alleles identified through Genome Wide Association Studies (GWAS).
Collapse
Affiliation(s)
- Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2049
|
Cavalli M, Pan G, Nord H, Wadelius C. Looking beyond GWAS: allele-specific transcription factor binding drives the association of GALNT2 to HDL-C plasma levels. Lipids Health Dis 2016; 15:18. [PMID: 26817450 PMCID: PMC4728761 DOI: 10.1186/s12944-016-0183-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 01/15/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Plasma levels of high-density lipoprotein cholesterol (HDL-C) have been associated to cardiovascular disease. The high heritability of HDL-C plasma levels has been an incentive for several genome wide association studies (GWASs) which identified, among others, variants in the first intron of the GALNT2 gene strongly associated to HDL-C levels. However, the lead GWAS SNP associated to HDL-C levels in this genomic region, rs4846914, is located outside of transcription factor (TF) binding sites defined by chromatin immunoprecipitation followed by DNA sequencing (ChIP-seq) experiments in the ENCODE project and is therefore unlikely to be functional. In this study we apply a bioinformatics approach which rely on the premise that ChIP-seq reads can identify allele specific binding of a TF at cell specific regulatory elements harboring allele specific SNPs (AS-SNPs). EMSA and luciferase assays were used to validate the allele specific binding and to test the enhancer activity of the regulatory element harboring the AS-SNP rs4846913 as well as the neighboring rs2144300 which are in high LD with rs4846914. FINDINGS Using luciferase assays we found that rs4846913 and the neighboring rs2144300 displayed allele specific enhancer activity. We propose that an inhibitor binds preferentially to the rs4846913-C allele with an inhibitory boost from the synergistic binding of other TFs at the neighboring SNP rs2144300. These events influence the transcription level of GALNT2. CONCLUSIONS The results suggest that rs4846913 and rs2144300 drive the association to HDL-C plasma levels through an inhibitory regulation of GALNT2 rather than the reported lead GWAS SNP rs4846914.
Collapse
Affiliation(s)
- Marco Cavalli
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Gang Pan
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Helena Nord
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Claes Wadelius
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
2050
|
Lamparter D, Marbach D, Rueedi R, Kutalik Z, Bergmann S. Fast and Rigorous Computation of Gene and Pathway Scores from SNP-Based Summary Statistics. PLoS Comput Biol 2016; 12:e1004714. [PMID: 26808494 PMCID: PMC4726509 DOI: 10.1371/journal.pcbi.1004714] [Citation(s) in RCA: 225] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 12/17/2015] [Indexed: 12/17/2022] Open
Abstract
Integrating single nucleotide polymorphism (SNP) p-values from genome-wide association studies (GWAS) across genes and pathways is a strategy to improve statistical power and gain biological insight. Here, we present Pascal (Pathway scoring algorithm), a powerful tool for computing gene and pathway scores from SNP-phenotype association summary statistics. For gene score computation, we implemented analytic and efficient numerical solutions to calculate test statistics. We examined in particular the sum and the maximum of chi-squared statistics, which measure the strongest and the average association signals per gene, respectively. For pathway scoring, we use a modified Fisher method, which offers not only significant power improvement over more traditional enrichment strategies, but also eliminates the problem of arbitrary threshold selection inherent in any binary membership based pathway enrichment approach. We demonstrate the marked increase in power by analyzing summary statistics from dozens of large meta-studies for various traits. Our extensive testing indicates that our method not only excels in rigorous type I error control, but also results in more biologically meaningful discoveries. Genome-wide association studies (GWAS) typically generate lists of trait- or disease-associated SNPs. Yet, such output sheds little light on the underlying molecular mechanisms and tools are needed to extract biological insight from the results at the SNP level. Pathway analysis tools integrate signals from multiple SNPs at various positions in the genome in order to map associated genomic regions to well-established pathways, i.e., sets of genes known to act in concert. The nature of GWAS association results requires specifically tailored methods for this task. Here, we present Pascal (Pathway scoring algorithm), a tool that allows gene and pathway-level analysis of GWAS association results without the need to access the original genotypic data. Pascal was designed to be fast, accurate and to have high power to detect relevant pathways. We extensively tested our approach on a large collection of real GWAS association results and saw better discovery of confirmed pathways than with other popular methods. We believe that these results together with the ease-of-use of our publicly available software will allow Pascal to become a useful addition to the toolbox of the GWAS community.
Collapse
Affiliation(s)
- David Lamparter
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Daniel Marbach
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Rico Rueedi
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Zoltán Kutalik
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- * E-mail: ;
| | - Sven Bergmann
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- * E-mail: ;
| |
Collapse
|